You are on page 1of 24

Clinical Pharmacokinetics

and Pharmacodynamics
Larry A. Bauer

1
KEY CONCEPTS
Clinical pharmacokinetics is the discipline that describes
the absorption, distribution, metabolism, and elimination of
e|CHAPTER 5
identified, including CYP1A2, CYP2C9, CYP2C19, CYP2D6,
CYP2E1, and CYP3A4.
drugs in patients requiring drug therapy. 11 Membrane transporters are protein molecules concerned
2 Clearance is the most important pharmacokinetic parameter with the active transport of drugs across cell membranes.
because it determines the steady-state concentration for a The importance of transport proteins in drug bioavailability,
given dosage rate. Physiologically, clearance is determined elimination, and distribution is continuing to evolve.
by blood flow to the organ that metabolizes or eliminates A principal transport protein involved in the movement
the drug and the efficiency of the organ in extracting the of drugs across biologic membranes is P-glycoprotein.
drug from the bloodstream. P-glycoprotein is present in many organs, including the
3 The volume of distribution is a proportionality constant gastrointestinal (GI) tract, liver, and kidney. Other transport
that relates the amount of drug in the body to the serum protein families include the organic cation transporters,
concentration. The volume of distribution is used to the organic anion transporters, and the organic anion
calculate the loading dose of a drug that will immediately transporting polypeptides.
achieve a desired steady-state concentration. The value of 12 When deciding on initial doses for drugs that are renally
the volume of distribution is determined by the physiologic eliminated, the patient’s renal function should be assessed.
volume of blood and tissues and how the drug binds in A common, useful way to do this is to measure the patient’s
blood and tissues. serum creatinine concentration and convert this value
4 Half-life is the time required for serum concentrations to into an estimated creatinine clearance (CLcr est). For drugs
decrease by one-half after absorption and distribution are that are eliminated primarily by the kidney (≥60% of the
complete. It is important because it determines the time administered dose), some agents will need minor dosage
required to reach steady state and the dosage interval. Half- adjustments for CLcr est between 30 and 60 mL/min (0.50 and
life is a dependent kinetic variable because its value depends 1.00 mL/s), moderate dosage adjustments for CLcr est between
on the values of clearance and volume of distribution. 15 and 30 mL/min (0.25 and 0.50 mL/s), and major dosage
adjustments for CLcr est less than 15 mL/min (0.25 mL/s).
5 The fraction of drug absorbed into the systemic circulation
Supplemental doses of some medications also may be
after extravascular administration is defined as its
needed for patients receiving hemodialysis if the drug is
bioavailability.
removed by the artificial kidney or for patients receiving
6 Most drugs follow linear pharmacokinetics, whereby steady- hemoperfusion if the drug is removed by the hemofilter.
state serum drug concentrations change proportionally with
long-term daily dosing. 13 When deciding on initial doses for drugs that are hepatically
eliminated, the patient’s liver function should be assessed.
7 Some drugs do not follow the rules of linear The Child-Pugh score can be used as an indicator of a
pharmacokinetics. Instead of steady-state drug patient’s ability to metabolize drugs that are eliminated by
concentration changing proportionally with the dose, serum the liver. In the absence of specific pharmacokinetic dosing
concentration changes more or less than expected. These guidelines for a medication, a Child-Pugh score equal to 8
drugs follow nonlinear pharmacokinetics. or 9 is grounds for a moderate decrease (∼25%) in the initial
8 Pharmacokinetic models are useful to describe data sets, to daily drug dose for agents that are metabolized primarily
predict serum concentrations after several doses or different hepatically (≥60%), and a score of 10 or greater indicates
routes of administration, and to calculate pharmacokinetic that a significant decrease in the initial daily dose (∼50%) is
constants such as clearance, volume of distribution, and required for drugs that are metabolized mostly hepatically.
half-life. The simplest case uses a single compartment to
14 For drugs that exhibit linear pharmacokinetics, steady-
represent the entire body.
state drug concentration (Css) changes proportionally with
9 Factors to be taken into consideration when deciding on dose (D). To adjust a patient’s drug therapy, a reasonable
the best drug dose for a patient include age, gender, weight, starting dose is administered for an estimated three to five
ethnic background, other concurrent disease states, and half-lives. A serum concentration is obtained, assuming that
other drug therapy. it will reflect Css. Independent of the route of administration,
10 Cytochrome P450 is a generic name for the group of the new dose (Dnew) needed to attain the desired Css(Css,new) is
enzymes that are responsible for most drug metabolism calculated as Dnew = Dold(Css,new/Css,old), where Dold and Css,old are
oxidation reactions. Several P450 isozymes have been the old dose and old Css, respectively.
51
Copyright © 2014 McGraw-Hill Education. All rights reserved.
52

15 If it is necessary to determine the pharmacokinetic constants
eTable 5-1 Selected Therapeutic Ranges
for a patient to individualize his or her dose, a small
Drug Therapeutic Range
pharmacokinetic evaluation is conducted in the individual.
Additionally, Bayesian computer programs that aid in the Digoxin 0.5–2 ng/mL or mg/L
0.6–2.6 nmol/L
individualization of therapy are available for many different
drugs. Lidocaine 1.5–5 mcg/mL or mg/L
6.4–21 μmol/L

16 Pharmacodynamics is the study of the relationship between
Procainamide/N-acetylprocainamide 10–30 mcg/mL or mg/L
the concentration of a drug and the response obtained (total) 42–127 μmol/L
SECTION

in a patient. If pharmacologic effect is plotted against


concentration for most drugs, a hyperbola results with an Quinidine 2–5 mcg/mL or mg/L
asymptote equal to the maximum attainable effect. 6–15 μmol/L
Amikacina 20–30 mcg/mL or mg/L (peak)
34–51 μmol/L (peak)
  

<5 mcg/mL or mg/L (trough)


1 CLINICAL PHARMACOKINETICS <9 μmol/L (trough)

AND PHARMACODYNAMICS: Gentamicin, tobramycin, netilmicina 5–10 mcg/mL or mg/L (peak)


10–21 μmol/L (peak)
Foundation Issues

INTRODUCTION <2 mcg/mL or mg/L (trough)


<4 μmol/L (trough)
Pharmacokinetic concepts have been used successfully by phar- Vancomycin 20–40 mcg/mL or mg/L (peak)
macists to individualize patient drug therapy for about a quarter 14–28 μmol/L (peak)
century. Pharmacokinetic consultant services and individual clini- 5–15 mcg/mL or mg/L (trough)b
3–10 μmol/L (trough)b
cians routinely provide patient-specific drug-dosing recommenda-
tions that increase the efficacy and decrease the toxicity of many Chloramphenicol 10–20 mcg/mL or mg/L
31–62 μmol/L
medications. Laboratories routinely measure patient serum or
plasma samples for many drugs, including antibiotics (e.g., ami- Lithium 0.6–1.4 mEq/L
0.6–1.4 mmol/L
noglycosides and vancomycin), theophylline, antiepileptics (e.g.,
Carbamazepine 4–12 mcg/mL or mg/L
phenytoin, carbamazepine, valproic acid, phenobarbital, and etho-
17–51 μmol/L
suximide), methotrexate, lithium, antiarrhythmics (e.g., lidocaine
Ethosuximide 40–100 mcg/mL or mg/L
and digoxin), and immunosuppressants (e.g., cyclosporine and
283–708 μmol/L
tacrolimus). Combined with a knowledge of the disease states and
Lamotrigine 2–20 mcg/mL
conditions that influence the disposition of a particular drug, kinetic
8–80 μmol/L
concepts can be used to modify doses to produce serum drug con-
Oxcarbazepine (as monohydroxy 3–35 mcg/mL
centrations that result in desirable pharmacologic effects without
derivative)
unwanted side effects. This narrow range of concentrations within
Phenobarbital 15–40 mcg/mL or mg/L
which the pharmacologic response is produced and adverse effects
65–172 μmol/L
prevented in most patients is defined as the therapeutic range of
Phenytoin/Fosphenytoin 10–20 mcg/mL or mg/L
the drug. eTable 5-1 lists the therapeutic ranges for commonly used
40–79 μmol/L
medications.
Primidone 5–12 mcg/mL or mg/L
Although most individuals experience favorable effects with
23–55 μmol/L
serum drug concentrations in the therapeutic range, the effects of a
Valproic acid 50–100 mcg/mL or mg/L
given serum concentration can vary widely among individuals. Cli-
347–693 μmol/L
nicians should never assume that a serum concentration within the
Theophylline 10–20 mcg/mL or mg/L
therapeutic range will be safe and effective for every patient. The
56–111 μmol/L
response to the drug, such as the number of seizures a patient experi-
Cyclosporine (blood) 150–400 ng/mL or mcg/L
ences while taking an antiepileptic agent, should always be assessed 125–333 nmol/L
when serum concentrations are measured.
Throughout this chapter, abbreviations for various pharmaco- a
Using a multiple dose per day conventional dosage schedule.
b
For patients with pneumonia or other life-threatening infections due to multi-
kinetic parameters are used frequently. eTable 5-2 lists commonly drug resistant bacteria trough concentrations as high as 15–20 mcg/mL or mg/L
used abbreviations. (10–14 μmol/L) have been recommended.

CLINICAL PHARMACOKINETIC The vascular system generally provides the “transportation”


CONCEPTS for the drug molecule to its site of activity. After the drug reaches
the systemic circulation, it can leave the vasculature and penetrate
1 Clinical pharmacokinetics is the discipline that describes the the various tissues or remain in the blood. If the drug remains
absorption, distribution, metabolism, and elimination of drugs in in the blood, it may bind to endogenous protein, such as albumin
patients requiring drug therapy. When a drug is administered extra- and α1-acid glycoprotein. This binding usually is reversible, and an
vascularly to patients, it must be absorbed across biologic mem- equilibrium is created between protein-bound drug and unbound
branes to reach the systemic circulation. If the drug is given orally, drug. Unbound drug in the blood provides the driving force for dis-
the drug molecules must pass through the gastrointestinal (GI) tract tribution of the agent to body tissues. If unbound drug leaves the
wall into capillaries. For transdermal patches, the drug must pen- bloodstream and distributes to tissue, it may become tissue-bound,
etrate the skin to enter the vascular system. In general, the pharma- it may remain unbound in the tissue, or if the tissue can metabolize
cologic effect of the drug is delayed when it is given extravascularly or eliminate the drug, it may be rendered inactive and/or eliminated
because time is required for the drug to be absorbed into the vas- from the body. If the drug becomes tissue-bound, it may bind to
cular system. the receptor that causes its pharmacologic or toxic effect or to a
Copyright © 2014 McGraw-Hill Education. All rights reserved.
53
eTable 5-2 Pharmacokinetic Abbreviations
Abbreviation Definition
CL Clearance
k0 IV infusion rate
CSS Steady-state concentration
D Dose

e|CHAPTER  
C
τ Dosage interval
F Fraction of drug absorbed into the systemic circulation
Q Blood flow
E Extraction ratio
fb Fraction of drug in the blood that is unbound
Clint Intrinsic clearance
Css, u Steady-state concentration of unbound drug
t 5
VD Volume of distribution
eFigure 5-1  Typical serum concentration-time curve following

Clinical Pharmacokinetics and Pharmacodynamics


LD Loading dose
MD Maintenance dose a continuous IV infusion.
t1/2 Half-life
k Elimination rate constant
ka Absorption rate constant
example, if a drug dose were doubled from 300 to 600 mg daily, the
α Distribution rate constant patient’s serum drug concentration would double.
β Terminal rate constant When a drug is given by continuous IV infusion, serum con-
t’ Postinfusion time centrations increase until an equilibrium is established between the
T Duration of infusion drug dosage rate and the rate of drug elimination. At that point, the
AUC Area under serum or blood concentration-versus- rate of drug administration equals the rate of drug elimination, and
time curve the serum concentrations remain constant (eFig. 5-1). For example,
Vmax Maximum rate of drug metabolism if a patient were receiving a continuous IV infusion of theophylline
at 40 mg/h, the theophylline serum concentration would increase
Km Serum concentration at which the rate of metabolism
equals Vmax/2 until the patient’s body was eliminating theophylline at 40 mg/h.
Cmax Maximum serum or blood concentration
When serum drug concentrations reach a constant value, steady
state is achieved.
Cmin Minimum serum or blood concentration
If the drug is given at intermittent dosage intervals, such as
DR Dosage rate 250 mg every 6 hours, steady state is achieved when the serum-­
P-gp P-glycoprotein concentration-versus-time curves for each dosage interval are
superimposable. The amount of drug eliminated during the dosage
interval equals the dose.

nonspecific binding site that causes no effect. Again, tissue binding Bioavailability and Bioequivalence
is usually reversible, so that the tissue-bound drug is in equilibrium
5 When drugs are administered extravascularly, drug molecules
with the unbound drug in the tissue.
must be released from the dosage form (dissolution) and pass
Certain organs—such as the liver, GI tract wall, and lung—­
through several biologic barriers before reaching the vascular sys-
possess enzymes that metabolize drugs. The resulting metabolite
tem (absorption). The fraction of drug absorbed into the systemic
may be inactive or have a pharmacologic effect of its own. The
circulation (F) after extravascular administration is defined as its
blood also contains esterases, which cleave ester bonds in drug mol-
bioavailability and can be calculated after single IV and extravas-
ecules and generally render them inactive.
cular doses as1
Drug metabolism usually occurs in the liver through one or
both of two types of reactions. Phase I reactions generally make the Div(AUC0 – ∞)
drug molecule more polar and water soluble so that it is prone to F=
D(AUCiv,0 – ∞)
elimination by the kidney. Phase I modifications include oxidation,
hydrolysis, and reduction. Phase II reactions involve conjugation to where D and Div are the extravascular and IV doses, respectively, and
form glucuronides, acetates, or sulfates. These reactions generally AUCiv,0-∞ and AUC0-∞ are the IV and extravascular areas under the
inactivate the pharmacologic activity of the drug and may make it serum- or blood-concentration-versus-time curves, respectively, from
more prone to elimination by the kidney. time zero to infinity. The AUC represents the body’s total exposure to
Other organs have the ability to eliminate drugs or metabolites the drug and is a function of the fraction of the drug dose that enters
from the body. The kidney can excrete drugs by glomerular filtra- the systemic circulation via the administered route and clearance
tion or by such active processes as proximal tubular secretion. Drugs (eFig. 5-2). When F is less than 1 for a drug administered extravascu-
also can be eliminated via bile produced by the liver or air expired larly, either the dosage form did not release all the drug contained in
by the lungs. it, or some of the drug was eliminated or destroyed (by stomach acid
or other means) before it reached the systemic circulation.
When the extravascular dose is administered orally, part of
Linear Pharmacokinetics the dose may be metabolized by enzymes or removed by transport
6 Most drugs follow linear pharmacokinetics; serum drug con- proteins contained in the GI tract wall or liver before it reaches
centrations change proportionally with long-term daily dosing. For the systemic circulation.2,3 This occurs commonly when drugs
Copyright © 2014 McGraw-Hill Education. All rights reserved.
54
16 where F is the fraction of dose absorbed into the systemic vascu-
lar system. The average Css over the dosage interval is the Css that
14 would have occurred had the same dose been given as a continuous
IV infusion (e.g., 300 mg every 6 hours would produce an average
12
Css equivalent to the actual Css produced by a continuous infusion
Concentration (mcg/mL)

10
administered at a rate of 50 mg/h).
Physiologically, clearance is determined by (a) blood flow (Q)
8 to the organ that metabolizes (liver) or eliminates (kidney) the drug
and (b) the efficiency of the organ in extracting the drug from the
SECTION

6 bloodstream.5 Efficiency is measured using an extraction ratio (E),


AUC calculated by subtracting the concentration in the blood leaving the
4 extracting organ (Cout) from the concentration in the blood entering
the organ (Cin) and then dividing the result by Cin:
  

2
Cin – Cout
1 0
0 5 10 15 20 25
E=
Cin
Time (h)
Clearance for that organ is calculated by taking the product of Q and
Foundation Issues

eFigure 5-2  Area under the concentration-versus-time curve E (CL = QE). For example, if liver blood flow equals 1.5  L/min,
(AUC) after the administration of an extravascular dose. The and the drug’s extraction ratio is 0.33, hepatic clearance equals
AUC is a function of the fraction of drug dose that enters the 0.5  L/min. Total clearance is computed by summing all the indi-
systemic circulation and clearance. AUCs measured after IV and vidual organ clearance values. Clearance changes occur in patients
extravascular doses can be used to determine bioavailability for when the blood flow to extracting organs changes or when the
the extravascular dose. extraction ratio changes. Vasodilators such as hydralazine and
nifedipine increase liver blood flow, whereas chronic heart failure
(CHF) and hypotension can decrease hepatic blood flow. Extrac-
tion ratios can increase when enzyme inducers increase the amount
have a high liver extraction ratio or are subject to GI tract wall of drug-metabolizing enzyme. Extraction ratios may decrease if
metabolism because, after oral administration, the drug must pass enzyme inhibitors inhibit drug-metabolizing enzymes or necrosis
through the GI tract wall and into the portal circulation of the liver. causes loss of parenchyma.
Transport proteins are also present in the GI tract wall that can
actively pump drug molecules that already have been absorbed
back into the lumen of the GI tract. P-glycoprotein (P-gp) is the Intrinsic Clearance
primary transport protein that interferes with drug absorption by The extraction ratio also can be thought of in terms of the unbound
this mechanism. For example, if an orally administered drug is fraction of drug in the blood (fb), the intrinsic ability of the extract-
100% absorbed from the GI tract but has a hepatic extraction ratio ing organ to clear unbound drug from the blood (CLint), and blood
of 0.75, only 25% of the original dose enters the systemic circula- flow to the organ (Q):6,7
tion. This first-pass effect through the liver and/or GI tract wall is fb(CLint)
avoided when the drug is given by other routes of administration. E=
Q + fb(CLint)
The computation of F does not separate loss of oral drug metabo-
lized by the first-pass effect and drug not absorbed by the GI tract. By substituting this equation for E, the clearance equation
Special techniques are needed to determine the fraction of drug becomes
absorbed orally for drugs with high liver extraction ratios or sub-
stantial gut wall metabolism. Q[ f b(CLint)]
CL =
Two different dosage forms of the same drug are considered to Q + f b(CLint)
be bioequivalent when the AUC0-∞, maximum serum or blood con-
centrations (Cmax), and the times that Cmax occurs (tmax) are neither Clearance changes will occur when blood flow to the clear-
clinically nor statistically different. When this occurs, the serum- ing organ changes (in conditions where blood flow is reduced,
concentration-versus-time curves for the two dosage forms should e.g., shock and CHF, or where blood flow is increased, e.g.,
be superimposable and identical. Bioequivalence studies have administration of medications, such as vasodilators, and resolu-
become very important as expensive drugs become available in tion of shock or CHF), binding in the blood changes (e.g., if the
less costly generic form. Most bioequivalence studies involve 18 to concentration of binding proteins is low or highly protein-bound
25 healthy adults who are given the brand-name product and the drugs are displaced), or intrinsic clearance of unbound drug
generic product in a randomized, crossover study design. changes (e.g., when metabolizing enzymes are induced or inhib-
ited by other drug therapy or functional organ tissue is destroyed
by disease processes).
Clearance If CLint is large (enzymes have a high capacity to metabolize
2 Clearance (CL) is the most important pharmacokinetic param- the drug), the product of fb and CLint is much larger than Q. When
eter because it determines the steady-state drug concentration (Css) fb(CLint) is much greater than Q, the sum of Q and fb(CLint) in the
for a given dosage rate. When a drug is given at a continuous IV denominator of the clearance equation almost equals fb(CLint):
infusion rate equal to k0, the Css is determined by the quotient of k0
and CL (Css = k0/CL). If the drug is administered as individual doses fb(CLint) ≈ Q + fb(CLint)
(D) at a given dosage interval (τ), the average Css over the dosage
interval is given by the equation4 Substituting this expression in the denominator of the clear-
ance equation and canceling common terms leads to the following
F(D/τ) expression for drugs with a large CLint: CL ≈ Q. In this case, clear-
Css =
CL ance of the drug is equal to blood flow to the organ; such drugs
Copyright © 2014 McGraw-Hill Education. All rights reserved.
55
are called high-clearance drugs and have large extraction ratios. that clinicians may increase the dose of a low-clearance drug after
Propranolol, verapamil, morphine, and lidocaine are examples of a protein-binding displacement interaction because Css decreased.
high-clearance drugs. High-clearance drugs such as these typically Because Css,u and the pharmacologic effect do not change, the dose
exhibit high first-pass effects when administered orally. should remain unaltered. Plasma protein binding decreases occur
If CLint is small (enzymes have a limited capacity to metabo- commonly in patients taking phenytoin. Low albumin concentra-
lize the drug), Q is much larger than the product of fb and CLint. tions (as in trauma or pregnant patients), high concentrations of
When Q is much greater than fb(CLint), the sum of Q and fb(CLint) in endogenous plasma protein-binding displacers (as with high con-

e|CHAPTER  
the denominator of the clearance equation becomes almost equal to centrations of bilirubin), or plasma protein-binding drug interac-
Q: Q ≈ Q + fb(CLint). Substituting this expression in the denomina- tions (as with concomitant therapy with valproic acid) can result
tor of the clearance equation and canceling common terms leads in subtherapeutic total phenytoin concentrations. Despite this fact,
to the following expression for drugs with a small CLint: CL ≈ unbound phenytoin concentrations usually are within the therapeu-
fb(CLint). In this case, clearance of the drug is equal to the product tic range, and often the patient is responding appropriately to treat-
of the fraction unbound in the blood and the intrinsic ability of the ment. Thus, in these situations, unbound rather than total phenytoin
organ to clear unbound drug from the blood; such drugs are known serum concentrations should be monitored and used to guide future
as low-clearance drugs and have small extraction ratios. Warfarin,
theophylline, diazepam, and phenobarbital are examples of low-
therapeutic decisions.
5
clearance drugs.
Clearances for Different Routes of

Clinical Pharmacokinetics and Pharmacodynamics


As mentioned previously, the concentration of unbound drug
in the blood is probably more important pharmacologically than Elimination and Metabolic Pathways
the total (bound plus unbound) concentration. The unbound drug Clearances for individual organs can be computed if the excretion
in the blood is in equilibrium with the unbound drug in the tissues the organ produces can be obtained. For example, renal clearance
and reflects the concentration of drug at its site of action. There- can be calculated if urine is collected during a pharmacokinetic
fore, the pharmacologic effect of a drug is thought to be a function experiment. The patient empties his or her bladder immediately
of the concentration of unbound drug in the blood. The unbound before the dose is given. Subsequent urine production is collected
steady-state concentration (Css,u) can be calculated by multiplying until the last serum concentration (Clast) is obtained. Renal clearance
Css and fb: Css,u = Css fb. The effect that changes in Q, fb, and CLint (CLR) is computed by dividing the amount of drug excreted in the
have on Css,u and therefore on the pharmacologic response of a drug urine by AUC0–t,last. Biliary and other clearance values are computed
depends on whether a high- or low-clearance drug is involved. in a similar fashion.
Because CL = Q for high-clearance drugs, a change in fb or CLint Clearances also can be calculated for each metabolite that is
does not change CL or Css(Css = k0/CL). However, a change in formed from the parent drug. This computation is particularly useful
unbound drug fraction does alter Css,u(Css,u = fbCss), thereby affect- in drug-interaction studies to determine which metabolic pathway is
ing the pharmacologic response. Plasma protein-binding displace- stimulated or inhibited. In the following metabolic scheme, the par-
ment drug interactions can be very important clinically, but they ent drug (D) is metabolized into two different metabolites (M1, M2)
are also dangerous because the changes in Css,u are not reflected in that subsequently are eliminated by the kidney (M1R, M2R):
changes in Css for high-clearance drugs. Because laboratories usu-
ally measure only total concentrations (concentrations of unbound CLFM1 kidney
D M1 M1R
drug are difficult to determine), the interaction is hard to detect. If
CLint changes for high-clearance drugs, CL, Css, Css,u, and pharma- CLFM2
cologic response do not change. Changes in Q cause a change in
CL; changes in Css, Css,u, and drug response are indirectly propor- M2 kidney M2R
tional to changes in CL.
For low-clearance drugs, total clearance is determined by To compute the formation clearance of M1 and M2 (CLFM1,
unbound drug fraction and intrinsic clearance: CL = fb(CLint). CLFM2), urine would be collected for five or more half-lives after a
A change in Q does not change CL, Css, Css,u, or pharmacologic single dose or during a dosage interval at steady state. The amount
response. However, a change in fb or CLint does alter CL and Css (Css = of metabolite eliminated in the urine is then determined. The frac-
k0/CL). Changes in CLint will cause a proportional change in CL. tion of the dose (in moles, because the molecular weights of the
Changes in Css, Css,u, and drug response are indirectly propor- parent drug and metabolites are not equal) eliminated by each meta-
tional to changes in CL. Altering fb for low-clearance drugs pro- bolic pathway (fM1 = M1R/D and fM2 = M2R/D) can then be computed.
duces interesting results. A change in fb alters CL and Css (Css  = Formation clearance for each pathway can be calculated using the
k0/CL). Because CL and Css change in opposite directions with following equations: CLFM1 = fM1CLM and CLFM2 = fM2CLM, where
changes in fb, Css,u (Css,u = fbCss) and pharmacologic response do not CLM is the metabolic clearance for the parent drug.
change with alterations in the fraction of unbound drug in the blood.
For example, a low-clearance drug is administered to a patient until
steady-state is achieved: Volume of Distribution
3 The volume of distribution (VD) is a proportionality constant
CL = f b(CLint) that relates the amount of drug in the body to the serum concen-
k0 tration (amount in body = CVD). VD is used to calculate the loading
Css = dose (LD) of a drug that will immediately achieve a desired Css
CL
(LD = CssVD). However, in practice, the patient’s own VD is not
Suppose that another drug is administered to the patient that known at the time the loading dose is administered. In this case,
displaces the first drug from plasma-protein-binding sites and an average VD is assumed and used to calculate a loading dose.
doubles fb (fb now equals 2fb). CL doubles because of the protein- Because the patient’s VD is almost always different from the aver-
binding displacement [2CL = 2fb(CLint)], and Css decreases by one- age VD for the drug, a loading dose does not attain the calculated
half because of the change in clearance [½(Css) = k0/(2Cl)]. Css,u Css, but it ideally achieves a therapeutic concentration. As usual,
does not change because even though fb is doubled, Css decreased steady-state conditions are achieved in three to five half-lives for
by one-half (Css,u = fbCss). The potential for error in this situation is the drug.
Copyright © 2014 McGraw-Hill Education. All rights reserved.
56

Linear
C0 Michaelis–Menten

12

Css or AUC
Slope = −k
2.303
log C
SECTION

Nonlinear protein binding


or autoinduction
6
t1/2
  

1 Dose
t
Foundation Issues

eFigure 5-4  Relationship of dose and steady-state drug


eFigure 5-3  Calculation of the half-life of a drug following IV concentration (Css) or area under the concentration-versus-time
bolus dosing. curve (AUC) under linear and nonlinear conditions.

The numeric value for the volume of distribution is determined concentrations change more or less than expected (eFig. 5-4). One
by the physiologic volume of blood and tissues and how the drug explanation for the greater-than-expected increase in Css and AUC
binds in blood and tissues:8 after an increase in dose is that the enzymes responsible for the
metabolism or elimination of the drug may start to become satu-
VD = Vb + (fb/ft)Vt
rated. When this occurs, the maximum rate of metabolism (Vmax)
where Vb and Vt are the volumes of blood and tissues, respectively, for the drug is approached. This is called Michaelis-Menten kinet-
and fb and ft are the fractions of unbound drug in blood and tissues, ics. The serum concentration at which the rate of metabolism equals
respectively. Vmax/2 is Km. Practically speaking, Km is the serum concentration
at which nonproportional changes in Css and AUC start to occur
Half-Life when the dose is increased. The Michaelis-Menten constants (Vmax
and Km) determine the dosage rate (DR) needed to maintain a given
4 Half-life (t1/2) is the time required for serum concentrations to
Css: DR = VmaxCss/(Km + Css). Most drugs eliminated by the liver are
decrease by one-half after absorption and distribution are complete.
metabolized by enzymes but still appear to follow linear kinetics.
It takes the same amount of time for serum concentrations to drop
The reason for this disparity is that the therapeutic range for most
from 200 to 100 mg/L as it does for concentrations to decline from
drugs is well below the Km of the enzyme system that metabolizes
2 to 1 mg/L (eFig. 5-3).
the agent. The therapeutic range is higher than Km for some com-
Half-life is important because it determines the time required
monly used drugs. The average Km for phenytoin is about 4 mg/L
to reach steady state and the dosage interval. It takes approximately
(16 μmol/L). The therapeutic range for phenytoin is usually 10 to
three to five half-lives to reach steady-state concentrations during
20 mg/L (40  to 79 μmol/L). Most patients experience Michaelis-
continuous dosing. In three half-lives, serum concentrations are at
Menten kinetics while taking phenytoin.
∼90% of their ultimate steady-state values. Because most serum drug
assays have an ∼10% error, it is difficult to differentiate concentra-
tions that are within 10% of each other. For this reason, many clini- Nonlinear Protein Binding
cians consider concentrations obtained after three half-lives to be Css. Another type of nonlinear kinetics can occur if Css and AUC increase
Half-life is also used to determine the dosage interval for less than expected after an increase in dose of a low-clearance drug.
a drug. For example, it may be desirable to maintain maximum This usually indicates that plasma protein-binding sites are start-
steady-state concentrations at 20 mg/L and minimum steady-state ing to become saturated, so that fb increases with increases in the
concentrations at 10 mg/L. In this case, it would be necessary to dose (see eFig. 5-4). For a low-clearance drug, CL depends on the
administer the drug every half-life because the minimum desirable values of fb and CLint (CL = fbCLint). When a dosage increase takes
concentration is one-half the maximum desirable concentration. place, fb increases because nearly all plasma protein-binding sites
Half-life is a dependent kinetic variable because its value are occupied, and no binding sites are available. If fb increases,
depends on the values of CL and VD.8 The equation that describes CL increases, and Css increases less than expected with the dos-
the relationship among the three variables is t1/2 = 0.693VD/CL. age change (Css = k0/CL). However, Css,u increases proportionally
Changes in t1/2 can result from a change in either VD or CL; a change with the dose because Css,u depends on CLint for low-clearance drugs
in t1/2 does not necessarily indicate that CL has changed. Half-life (Css,u = k0/CLint). Valproic acid9 and disopyramide10 both follow satu-
can change solely because of changes in VD. The elimination rate rable protein-binding pharmacokinetics.
constant (k) is related to the half-life by the following equation: k =
0.693/t1/2. Both the half-life and elimination rate constant describe
how quickly serum concentrations decrease in the serum or blood.
Autoinduction
For some drugs, clearance increases as the dose or concentration of
the drug increases. In this situation, increasing the drug dose or con-
Nonlinear Pharmacokinetics centration increases the ability of the enzyme system to eliminate
Michaelis-Menten Kinetics the compound and to clear the drug from the body. This is usually
7 Some drugs do not follow the rules of linear pharmacokinetics. accomplished by inducing the enzyme system responsible for the
Instead of Css and AUC increasing proportionally with dose, serum metabolism of the drug, so that the intrinsic clearance of the drug
Copyright © 2014 McGraw-Hill Education. All rights reserved.
57
One compartment
A
k0,ka, or k A
1 log
IV bolus
residual −ka
Slope =
2.303

t
Two compartments 10

e|CHAPTER  
log C
k12 −k
k0,ka, or Slope =
2.303
1 2
IV bolus
k21
k10
5

t 1/2
eFigure 5-5  Visual representations of one- and two-compartment
drug-distribution models. 5
t

Clinical Pharmacokinetics and Pharmacodynamics


increases. Because the drug itself is causing the induction effect, this eFigure 5-6  Calculation of the half-life of a drug following oral,
process is called autoinduction. For some drugs, such as carbamaze- intramuscular, or other extravascular dosing route. Inset shows
pine,11 the autoinduction effect is continuous within the typical dos- calculation of the absorption rate constant (ka) using the method
age range, which produces a curve for the dose versus Css or AUC of residuals.
plot similar to nonlinear protein binding (see eFig. 5-4). Detailed
pharmacokinetic studies are conducted to differentiate between non-
linear protein binding and autoinduction when dose versus Css or or t1/2 and using the formulas given for the IV bolus case. At each
AUC plots systematically deviate below the linear line. time point in the absorption portion of the curve, the concentration
value from the extrapolated line is noted and called the extrapolated
Pharmacokinetic Models and Equations concentration. For each point, the actual concentration is subtracted
8 Pharmacokinetic models are useful to describe data sets, to pre- from the extrapolated concentration to compute the residual con-
dict serum concentrations after several doses or different routes of centration. When the residual concentrations are plotted on semi-
administration, and to calculate pharmacokinetic constants such logarithmic coordinates (see eFig. 5-6, inset), a line with y intercept
as CL, VD, and t1/2.12 Compartmental models depict the body as equal to A and slope equal to -ka/2.303 is obtained. When these val-
one or more discrete compartments to which a drug is distributed ues are calculated, they can be placed into the equation (C = Ae−kt -
and/or from which a drug is eliminated. The shape of the serum-­ Ae−kat, where A = FDka/[VD(ka - k)]) and used to compute the serum
concentration-versus-time curve determines the number of com- concentration at any time after the extravascular dose. The inter-
partments in the pharmacokinetic model and the equation used in cepts and rate constants also can be used to compute CL and VD:
computations (eFig. 5-5). First-order rate constants, known as micro- CL = FD/(A/k - A/ka) and VD = CL/k, where F is the fraction of the
constants, describe the rate of transfer from one compartment to dose absorbed into the systemic circulation.
another. Each compartment also has its own VD. For clinical dosage During a continuous IV infusion, the serum concentrations in
adjustment purposes using drug concentrations, a one-compartment a one-compartment model change according to the following func-
model is the most commonly used pharmacokinetic model. tion: C = (k0/CL)(1 - e−kt). If the infusion has been running for more
than three to five half-lives, the patient will be at steady state, and
One-Compartment Model CL can be calculated (CL = k0/Css). When the infusion is discontin-
The simplest case uses a single compartment to represent the entire ued, serum concentrations appear to decline in a straight line when
body (see eFig. 5-5). The drug enters the compartment by continu- plotted on semilogarithmic paper with a slope of -k/2.303. VD is
ous IV infusion (k0), absorption from an extravascular site with an computed by dividing CL by k (eFig. 5-7).
absorption rate constant of ka, or IV bolus (D). After an IV bolus,
serum concentrations decline in a straight line when plotted on
semilogarithmic coordinates (see eFig. 5-3). The slope of the line is
-k/2.303; t1/2 can be computed by determining the time required for
concentrations to decrease by one-half (t1/2 = 0.693/k). The equation DC infusion
that describes the data is C = (D/VD)e−kt. VD is calculated by divid- Css

ing the IV dose by the y intercept (the concentration at time zero,


C0) of the graph. CL is computed by taking the product of k and VD.
log C

Once VD and k are known, concentrations at any time after the dose
can be computed [C = (D/VD)e−kt].
When an extravascular dose is given, one-compartment-model Slope =
−k
serum concentrations rise during absorption, reach Cmax, then 2.303

decrease in a straight line with a slope equal to –k/2.303. The equa-


tion that describes the data is C = {(FDka)/[VD(ka – k)]}(e−kt – e−kat),
where F is the fraction of the dose absorbed into the systemic circu- t
lation. The absorption rate constant (ka) is obtained using the method 3–5 t 1/2
of residuals.
The method of residuals is used to obtain the individual rate eFigure 5-7  Achievement of steady-state serum concentrations
constants (eFig. 5-6). A is determined by extrapolating the termi- after three to five half-lives of a drug. Note the elimination phase
nal slope to the y axis; k can be obtained by calculating the slope after discontinuance of the infusion.
Copyright © 2014 McGraw-Hill Education. All rights reserved.
58
Multicompartment Model R
−α
After an IV bolus dose, serum concentrations often decline in 7 Slope =
2.303
two or more phases. During the early phases, the drug leaves the log
bloodstream by two mechanisms: (a) distribution into tissues and residual 3.5
3.5
(b) metabolism and/or elimination. Because the drug is leaving the
t1/2α
bloodstream through these two mechanisms, serum concentrations
decline rapidly. After tissues and blood are in equilibrium, only t
metabolism and elimination remove the drug from the blood. Dur-

log C
−β
SECTION

ing this terminal phase, serum concentrations decline more slowly. Slope =
2.303
The half-life is measured during the terminal phase by determining
the time required for concentrations to decline by one-half. 6
After an IV bolus dose, serum concentrations decrease as if the S
3
drug were being injected into a central compartment that not only
  

t1/2β
metabolizes and eliminates the drug but also distributes the drug to
1 one or more other compartments. Of these multicompartment mod-
els, the two-compartment model is encountered most commonly
t
(see eFig. 5-5). After an IV bolus injection, serum concentrations
Foundation Issues

DC infusion
decrease in two distinct phases, described by the equation
D(α – k21) –αt D(k21 – β ) –β t eFigure 5-9  Calculation of α and β half-lives following a steady-
C= e + e state infusion. Inset shows calculation of the distribution rate
V D1(α – β ) V D1(α – β )
constant (α) using the method of residuals.
or C = Ae−αt + Be−βt, where k21 is the first-order rate constant that
reflects the transfer of the drug from compartment 2 to compart-
ment 1, VD is the VD of compartment 1, A = D(α - k21)/[VD (α - β )]
1 1 If serum concentrations of a drug given as a continuous IV
and B = D(k21 - β )/[VD1(α - β )]. The rate constants α and β found
infusion decline in a biphasic manner after the infusion is discontin-
in the exponents of the equations describe the distribution and elimi-
ued, a two-compartment model describes the data set (eFig. 5-9).13,14
nation of the drug, respectively (eFig. 5-8). A and B are the y inter-
In this instance, the postinfusion concentrations decrease accord-
cepts of the lines that describe drug distribution and elimination,
ing to the equation C = Re−αt′ + Se−βt′, where t′ is the postinfusion
respectively, on the log concentration-versus-time plot.
time (t’ = 0 when infusion is discontinued), and R, S, α, and β are
The residual line is calculated as before using the method of
determined from the postinfusion concentrations using the method
residuals (see eFig. 5-8, inset). The terminal line is extrapolated to
of residuals with the y axis set at t′ = 0. R and S are used to compute
the y axis, and extrapolated concentrations are determined for each
A and B. A and B are the y intercepts that would have occurred had
time point. Because actual concentrations are greater in this case,
the total dose given during the infusion (D = k0T) been administered
residual concentrations are calculated by subtracting the extrapo-
as an IV bolus dose:
lated concentrations from the actual concentrations. When plotted
on semilogarithmic paper, the residual line has a y intercept equal RDα
A=
to A. The slope of the residual line is used to compute α (slope = k0(1 – e–αT )
-α/2.303). With the rate constants (α and β ) and the intercepts
SDβ
(A and B), concentrations can be calculated for any time after the IV B=
bolus dose (C = Ae−αt + Be−βt), or pharmacokinetic constants can be k0(1 – e–βT )
computed: CL = D/[(A/α) + (B/β )], VD,β = CL/β, VD,ss = {D[(A/α 2) +
(B/β 2)]}/[(A/α) + (B/β )]2. where T is the duration of infusion. Once A, B, α, and β are known,
the equations for an IV bolus are used to compute the pharma-
cokinetic constants. Often, when a drug is given as an IV bolus
or continuous IV infusion, a two-compartment model is used to
A describe the data, but when the same agent is given extravascu-
−α
5 Slope = larly, a one-compartment model applies.15 In this case, distribu-
2.303
log tion occurs during the absorption phase, so a distribution  phase
residual 2.5 is not observed.
t1/2α
Volumes of Distribution
t
in Multicompartment Models
Two different VD values are needed as proportionality constants for
log C

B 20 −β drugs that require multicompartment models to describe the serum-


Slope =
2.303 concentration-versus-time curve. The VD that is used to compute
10 the amount of drug in the body during the terminal (β ) portion of
the curve is called VD,β (amount of drug in body = VD,β C). During
t1/2β a continuous IV infusion at steady state, VD,ss is used to compute
the amount of drug in the body (amount of drug in body = VD,ssC).
VD,ss is also the VD that can be computed using the physiologic vol-
t umes of blood and tissues and the ratio of unbound drug in blood
to that in tissues [VD,ss = Vb + (fb/ft)Vt]. Because the value of VD,β
eFigure 5-8  Calculation of α and β half-lives following IV changes when CL changes, VD,ss should be used to indicate if drug
dosing. Inset shows calculation of the distribution rate constant distribution changes during pharmacokinetic or drug-interaction
(α) using the method of residuals. experiments.
Copyright © 2014 McGraw-Hill Education. All rights reserved.
59
Multiple Dosing and Steady-State Equations eTable 5-3A Cytochrome P450 Enzyme Family
Any of these compartmental equations can be used to determine and Selected Substrates
serum concentrations after multiple doses. The multiple-dosing CYP1A2 CYP2E1
factor (1 - e−nKτ)/(1 - e−Kτ), where n is the number of doses, K is Acetaminophen Enflurane
the appropriate rate constant, and τ is the dosage interval, is simply Caffeine Ethanol
Ondansetron Halothane
multiplied by each exponential term in the equation, substituting
Tacrine Isoflurane
the rate constant of each exponent for K. Time (t) is set at 0 at

e|CHAPTER  
Theophylline CYP3A4
the beginning of each dosage interval. For example, a single-dose R-warfarin Alfentanil
two-compartment IV bolus is calculated as follows: C = Ae−αt + Zileuton Alprazolam
Be−βt. Thus, the equation for a multiple-dose two-compartment IV CYP2C9 Astemizole
bolus is Candesartan Carbamazepine
Diclofenac Cyclosporine
1 – e–nατ 1 – e–nβτ Ibuprofen Diltiazem
C = Ae–αt + Be–βt Losartan Erythromycin
1 – e–ατ 1 – e–βτ
5
Naproxen Felodipine
A single-dose one-compartment IV bolus is calculated as C = Phenytoin Itraconazole
Tolbutamide Ketoconazole
(D/VD)e−kt. For a multiple-dose one-compartment IV bolus, the con-
Valsartan Lidocaine
centration is C = (D/VD)e−kt[(1 - e−nkτ)/(1 - e−kτ)].

Clinical Pharmacokinetics and Pharmacodynamics


S-warfarin Lovastatin
At steady state, the number of doses becomes large, e−nKτ CYP2C19 Midazolam
approaches zero, and the multiple-dosing factor equals 1/(1 - e−Kτ). Diazepam Nifedipine
Therefore, the steady-state versions of the equations are simpler Lansoprazole Quinidine
(S)-mephenytoin Simvastatin
than their multiple-dose counterparts: Tacrolimus
Nelfinavir
Verapamil
Ae–αt Be–βt Omeprazole
C= + Pantoprazole Ziprasidone
1–e –ατ
1 – e–βτ
Voriconazole
and CYP2D6
Carvedilol
(D/VD)e –kt
Codeine
C=
1 – e–kτ Debrisoquine
Dextromethorphan
for a steady-state two-compartment IV bolus and a steady-state one- Encainide
compartment IV bolus, respectively. Fluoxetine
Haloperidol
(S)-metoprolol
Use of Pharmacokinetic Concepts for Paroxetine
Propafenone
Individualization of Drug Therapy Risperidone
Thioridazine
9 Many factors must be taken into consideration when deciding
Venlafaxine
on the best drug dose for a patient. For example, the age of the
patient is important because the dose (in milligrams per kilogram)
for pediatric patients may be higher and for geriatric patients may
be lower than the typically prescribed dose for young adults. Gender
also can be a factor because male and female patients metabolize CYP2D6 oxidizes debrisoquine.16 These subsets of the CYP enzyme
and eliminate some drugs differently. Patients who are significantly family are also responsible for the metabolism of several other drugs
obese or cachectic also may require different drug doses because of (CYP2D6: many tricyclic antidepressants, codeine, (S)-metoprolol;
clearance and volume of distribution changes. Other drug therapy CYP2C19: most proton pump inhibitors, sertraline, voriconazole).
that could cause drug interactions needs to be considered. Disease CYP2C9, CYP2C19, and CYP2D6 isozymes appear to be under
states and conditions may alter the drug-dosage regimen for a genetic control. As a consequence, there are “poor metabolizers”
patient. Three disease states that deserve special mention are CHF, who have a defective mutant gene for the isozyme, cannot manu-
renal disease, and hepatic disease. Renal and hepatic diseases cause facture a fully functional isozyme, and therefore cannot metabolize
loss of organ function and decreased drug elimination and metabo- the drug substrate very well. “Extensive metabolizers” have the
lism. CHF causes decreased blood flow to organs that clear the drug standard gene for the isozyme and metabolize the drugs normally.
from the body. Poor metabolizers usually are a minority of the general population.
Many drug compounds are racemic mixtures of stereoiso- They may achieve toxic concentrations of a drug when usual doses
mers. In most cases, one of the isomers is more pharmacologically are prescribed for them or, if the active drug moiety is a metabo-
active than the other isomer, and each isomer may exhibit differ- lite, may fail to have any pharmacologic effect from the drug. The
ent pharmacokinetic properties. Warfarin, propranolol, verapamil, ethnic background of the patient can affect the likelihood that the
and ibuprofen are all racemic mixtures of stereoisomers. Some drug patient will be a poor metabolizer.16 For example, the incidence of
interactions inhibit or increase the elimination of only one stereo- poor metabolizers for CYP2D6 is ∼5% to 10% for whites and ∼0%
isomer. The importance of the drug interaction depends on which to 1% for Asians, whereas for CYP2C19, poor metabolizers make
isomer is affected. Other drugs, such as dextromethorphan, levo- up ∼3% to 6% of the white population and ∼20% of the Asian popu-
floxacin, and diltiazem, are composed of just one stereoisomer. lation. Approximately 7% of the whites are poor metabolizers for
10 Genetics also plays a role in drug metabolism. Cytochrome CYP2C9 substrates.
P450 is a generic term for the group of enzymes that are responsible Other cytochrome P450 isozymes have been isolated.16
for most drug metabolism oxidation reactions. Several cytochrome CYP1A2 is the enzyme that is responsible for the demethylation
P450 (CYP) isozymes have been identified that are responsible for of caffeine and theophylline; CYP2C9 metabolizes phenytoin,
the metabolism of many important drugs (eTable 5-3A). CYP2C19 tolbutamide, losartan, and ibuprofen; some antiretroviral protease
is responsible for aromatic hydroxylation of (S)-mephenytoin, and inhibitors, cyclosporine, nifedipine, lovastatin, simvastatin, and
Copyright © 2014 McGraw-Hill Education. All rights reserved.
60
atorvastatin are metabolized by CYP3A4; and ethanol is a sub- eTable 5-3B Membrane Transport Proteins
strate for CYP2E1. It is important to recognize that a drug may and Selected Substrates
be metabolized by more than one cytochrome P450 isozyme.
P-Glycoprotein (P-gp) OAT1B1
Although most tricyclic antidepressants are hydroxylated by
Sites: Intestinal enterocytes, kidney Site: Hepatocytes (sinusoidal)
CYP2D6, N-­demethylation probably is mediated by a combina- proximal tubule, hepatocytes Bosentan
tion of CYP2C19, CYP1A2, and CYP3A4. Acetaminophen appears (canalicular), brain endothelia Olmesartan
to be metabolized by both CYP1A2 and CYP2E1. The 4-hydroxy Alfentanil Repaglinide
metabolite of propranolol is produced by CYP2D6, but side-chain Aliskiren Statins
Ambrisentan Valsartan
oxidation of propranolol is probably a product of CYP2C19. The
SECTION

Atorvastatin OAT1
CYP3A enzyme family comprises ∼90% of the drug-metabolizing Azithromycin Sites: Kidney proximal tubule,
enzyme present in the intestinal wall but only ∼30% of the drug- Cetirizine placenta
metabolizing enzyme found in the liver. The remainder of hepatic Citalopram Acyclovir
Clopidogrel
drug-metabolizing enzyme is ∼20% for the CYP2C family, ∼13% Cephradine
Cyclosporine
  

for CYP1A2, ∼7% for CYP2E1, and ∼2% for CYP2D6. Ciprofloxacin
Daunorubicin
1
Methotrexate
Understanding which cytochrome P450 isozyme is responsible Dexamethasone Zidovudine
for the metabolism of a drug is extraordinarily useful in predict- Digoxin
Diltiazem OAT3
ing and understanding drug interactions. Some drug-metabolism Sites: Kidney proximal tubule,
Doxorubicin
Foundation Issues

inhibitors and inducers are highly selective for certain CYP iso- Erythromycin choroid plexus, brain endothelia
zymes.16  Quinidine is an extremely potent inhibitor of the CYP2D6 Etoposide Bumetanide
enzyme system;16 a single 50-mg dose of quinidine can change a Fexofenadine Cefaclor
Glyburide Ceftizoxime
rapid metabolizer of debrisoquine into a poor metabolizer. Cipro- Furosemide
Indinavir
floxacin and zileuton inhibit, whereas tobacco and marijuana smoke NSAIDs
Imatinib
induces, CYP1A2. Some drugs that are enzyme inhibitors are also Loperamide OCT1
substrates for that same enzyme system and appear to cause drug Loratadine Sites: Hepatocytes (sinusoidal),
interactions by being a competitive inhibitor. For example, eryth- Lovastatin intestinal enterocytes
romycin is both a substrate for and an inhibitor of CYP3A4. Obvi- Morphine Metformin
Nelfinavir Oxaliplatin
ously, if one knows that a new drug is metabolized by a given CYP Olanzapine OCT2
enzyme system, it is logical to assume that the new drug will exhibit Ondansetron Sites: Kidney proximal tubule,
drug interactions with the known inducers and inhibitors of that Paclitaxel neurons
CYP isozyme. Quinidine Amantadine
Raltegravir Amiloride
11 The importance of membrane transport proteins in drug
Ranolazine Metformin
bioavailability, elimination, and distribution is now better under- Risperidone Pindolol
stood.16–18 Membrane transporters are protein molecules con- Rifampin Procainamide
cerned with the active transport of drugs across cell membranes Ritonavir Ranitidine
(eTable 5-3B). This results in the transfer of drug molecules either Saquinavir
Tacrolimus
out of or into cells. Membrane transporters have been found in the Telaprevir
intestine, liver, kidney, and the blood-brain barrier. Verapamil
A principal transport protein involved in the movement of Vinblastine
drugs across biologic membranes is P-gp. P-gp is present in many Vincristine
organs, including the GI tract, liver, and kidney. If a drug is a sub- OAT, organic anion transporter; OCT, organic cation transporter; NSAIDs,
strate for P-gp, its oral absorption may be decreased when P-gp nonsteroidal antiinflammatory drugs.
transports drug molecules that have been absorbed back into the
GI tract lumen. In the liver, some drugs are transported by P-gp
from the blood into the bile, where the drug is eliminated by biliary
secretion. Similarly, some drugs eliminated by the kidney are trans-
Selection of Initial Drug Doses
ported from the blood into the urine by P-gp. Digoxin is a substrate 12 When deciding on initial doses for drugs that are eliminated
of P-gp. Other possible mechanisms for drug interactions are when renally, the patient’s renal function should be assessed. A com-
two drugs that are substrates for P-gp compete for transport by the mon, useful way to do this is to measure the patient’s serum cre-
protein and when a drug is an inhibitor or inducer of P-gp. Drug atinine concentration and convert this value into a CLcr est. Serum
interactions involving inhibition of P-gp decrease drug transporta- creatinine values alone should not be used to assess renal function
tion in these organs and potentially can increase GI absorption of an because they do not include the effects of age, body weight, or gen-
orally administered drug, decrease biliary secretion of the drug, or der. The Cockcroft-Gault equation19 is probably the most widely
decrease renal elimination of drug molecules. The drug interaction used method to estimate creatinine clearance (CLcr) (in milliliters
between amiodarone and digoxin probably involves all three of these per minute) in adults (age 18 years or older) who are within ∼30%
mechanisms; this explains why digoxin concentrations increase so of their ideal body weight and have stable renal function:
dramatically in patients receiving amiodarone. Many drugs that are (140 – age)BW
metabolized by CYP3A4 are also substrates for P-gp, and some of Men: CLcr est =
Scr × 72
the drug interactions attributed to inhibition of CYP3A4 may be a
result of decreased drug transportation by P-gp. Drug interactions 0.85(140 – age)BW
Women: CLcr est =
involving induction of P-gp have the opposite effect in these organs Scr × 72
and may decrease GI absorption of an orally administered drug,
increase biliary secretion of the drug, or increase renal elimination where BW is body weight (in kilograms), age is the patient’s age
of drug molecules. (in  years), 0.85 is a correction factor to account for lower mus-
Other membrane transporter families include the organic cation cle mass in women, and Scr is serum creatinine (in milligrams
transporters (OCT family), organic anion transporters (OAT family), per deciliter). For children, the following estimation equations
and the organic anion transporting polypeptides (OATP family). are available according to the age of the child20 age 0 to 1 year:
Copyright © 2014 McGraw-Hill Education. All rights reserved.
61
CLcr  est (in mL/min/1.73 m2) = (0.45 × Lt)/Scr; age 1 to 20 years: eTable 5-4 Theophylline Pharmacokinetic Parameters
CLcr  est (in mL/min/1.73 m2) = (0.55 × Lt)/Scr, where Lt is patient for Selected Disease States/Conditions
length in centimeters. (To use these equations, Scr expressed in
Mean Clearance Mean Dose
μmol/L must first be divided by 88.4 to obtain conventional units Disease State/Condition (mL/min/kg) (mg/kg/h)
of mg/dL. Conversion of CLcr to units of mL/s/m2 requires multi-
Children 1–9 years old 1.4 0.8
plication of CLcr expressed in milliliters per minute per 1.73 m2 by
Children 9–12 years old or adult 1.25 0.7
0.00963.) Other methods to determine CLcr est for obese adults21 and
smokers

e|CHAPTER  
patients with rapidly changing renal function22 are available. Creati-
Adolescents 12–16 years old or 0.9 0.5
nine is a by-product of muscle breakdown in the body, so none of
elderly smokers (>65 years)
these estimation methods work well in patients with muscle disease,
Adult nonsmokers 0.7 0.4
such as multiple sclerosis, or diseases that alter muscle mass, such
as cachexia, malnutrition, cancer, or spinal cord injury. Nomograms Elderly nonsmokers (>65 years) 0.5 0.3
that adjust initial doses according to a patient’s renal function are Decompensated CHF, cor 0.35 0.2
available for several drugs, including digoxin,23 vancomycin,24 and pulmonale, cirrhosis
the aminoglycoside antibiotics.25
For drugs that are eliminated primarily by the kidney (≥60% of
Mean volume of distribution = 0.5 L/kg.
CHF, chronic heart failure.
5
the administered dose), some agents will need minor dosage adjust- Data from Reference 52.

Clinical Pharmacokinetics and Pharmacodynamics


ments for CLcr est between 30 and 60 mL/min (0.50 and 1.00 mL/s),
moderate dosage adjustments for CLcr est between 15 and 30 mL/min
(0.25 and 0.50 mL/s), and major dosage adjustments for CLcr est less
than 15 mL/min (0.25 mL/s). Specific recommendations for dos- therapeutic and adverse effects, and drug serum concentrations are
age adjustments of other drugs for patients with renal disease are obtained to ensure that concentrations are appropriate and to adjust
available.26,27 Supplemental doses of some medications also may be doses, if necessary. The following computations illustrate the esti-
needed for patients receiving hemodialysis if the drug is removed by mated IV loading dose and the IV continuous infusion necessary
the artificial kidney or for patients receiving hemoperfusion if the to achieve a theophylline concentration of 10 mg/L (10 mcg/mL;
drug is removed by the hemofilter.27 55.5 μmol/L) for a 55-year-old man, weighing 70 kg (154 lb), with
13 A similar assessment of liver function should be made for liver cirrhosis (mean kinetic parameters obtained from eTable 5-4):
drugs that are metabolized hepatically. Unfortunately, there is no VD = (0.5 L/kg)(70 kg) = 35 L
single test that can estimate liver drug-metabolism capacity accu- LD = CssVD = (10 mg/L)(35 L)
rately, and those that are used do not always prove accurate. High = 350 mg theophylline infused over 20 to 30 min
aminotransferase (aspartate aminotransferase [AST] and alanine (0.35 mL/min/kg)(70 kg)(60 min/h)
aminotransferase [ALT]) and alkaline phosphatase concentrations CL(in L/h) =
1000 mL/L
usually indicate acute hepatic cellular damage and do not establish = 1.5 L/h
poor liver drug metabolism reliably. Abnormal values for three tests k0 = CssCL = (10 mg/L)(1.5 L/h)
that usually indicate that drugs will be metabolized poorly by the = 15 mg/h of theophylline to begin after loading dose
liver are high serum bilirubin concentration, low serum albumin con- is given
centration, and a prolonged prothrombin time. Bilirubin is metabo-
lized by the liver, and albumin and clotting factors are manufactured If theophylline is to be given as the aminophylline salt
by the liver, so aberrant values for all three of these tests are a more form, each dose would need to be changed to reflect the fact that
reliable indicator of abnormal liver drug metabolism. The Child- ­aminophylline contains only 85% theophylline (LD = 350 mg of
Pugh score,28 a widely used clinical classification for liver disease ­theophylline/0.85 = 410 mg of aminophylline infused over 20 to
that incorporates clinical signs and symptoms (ascites and hepatic 30 minutes, k0 = 15 mg/h of theophylline/0.85 = 18 mg/h of amino-
encephalopathy), in addition to these three laboratory tests, can be phylline to begin after loading dose is given).
used as an indicator of a patient’s ability to metabolize drugs that are Heart failure is often overlooked as a disease state that can alter
eliminated by the liver. A score in excess of 10 suggests very poor drug disposition. Severe heart failure decreases cardiac output and
liver function. As a general rule, patients with cirrhosis have the most therefore reduces liver blood flow. Theophylline,29 lidocaine,30 and
severe decreases in liver drug metabolism. Patients with acute or drugs with high extraction ratios are compounds whose clearance
chronic hepatitis often retain relatively normal or slightly decreased declines with decreased liver blood flow. Initial dosages of these
hepatic drug-metabolism capacity. In the absence of specific phar- drugs should be reduced in patients with moderate to severe heart
macokinetic dosing guidelines for a medication, a Child-Pugh score failure (New York Heart Association class III or IV) by 25% to 50%
equal to 8 to 9 is grounds for a moderate decrease (∼25%) in initial until steady-state concentrations and response can be determined.
daily drug dose for agents that are metabolized primarily (≥60%)
hepatically, and a score of 10 or greater indicates that a significant Use of Steady-State Drug
decrease in initial daily dose (∼50%) is required for drugs that are
metabolized mostly by the liver. As in any patient with or without Concentrations
liver dysfunction, initial doses are meant as starting points for dosage 14 Serum drug concentrations are readily available to clinicians to
titration based on patient response and avoidance of adverse effects. use as guides for the individualization of drug therapy. The thera-
Because there are no good markers of liver function, clinicians peutic ranges for several drugs have been identified, and it is likely
have come to rely on pharmacokinetic parameters derived in vari- that new drugs also will be monitored using serum concentrations.
ous patient populations to compute initial doses of drugs that are Although several individualization methods have been advocated
eliminated hepatically. eTable 5-4 contains average pharmacokinetic for specific drugs, one simple, reliable method is used commonly.
parameters for theophylline in several disease states. Initial doses of For drugs that exhibit linear pharmacokinetics, Css changes pro-
many liver-metabolized drugs are computed by determining which portionally with the dose. To adjust a patient’s drug therapy, a rea-
disease states and/or conditions the patient has that are known to sonable starting dose is administered for an estimated three to five
alter the kinetics of the drug and by using these average pharmacoki- half-lives. A serum concentration is obtained, assuming that it will
netic constants to calculate doses. The patient is then monitored for reflect Css. Independent of the route of administration, the new dose
Copyright © 2014 McGraw-Hill Education. All rights reserved.
62
(Dnew) needed to attain the desired Css (Css,new) is calculated: Dnew = 10
C max,ss
Dold(Css,new/Css,old), where Dold and Css,old are the old dose and old Css,
respectively. To use this method, Css,old must reflect steady-state con- t1/2

Concentration (mcg/mL)
ditions. Often patients are noncompliant with regard to their drug
dosage and therefore are not at steady state. This occurs not only
in outpatients but also in hospital inpatients. Inpatients can spit out 1
oral doses or alter the infusion rates on IV pump rates after the nurse Cmin,ss

Cmin,ss (extrapolated)
leaves the hospital room. Doses also can be missed if the patient
is absent from his or her room at the time medications are to be
SECTION

administered. If Css,old is much larger or smaller than expected for


the Dold the patient is taking, one should suspect noncompliance and 0.1
repeat the serum concentration determination after another three to 0 1 2 3 4 5 6 7 8 9 10
five half-lives or change the patient’s dose cautiously and monitor Time (h)
  

for signs of toxicity or lack of effect.


eFigure 5-10  When a patient has received enough doses to be
1 Measurement of Pharmacokinetic
at steady state, steady-state maximum (Cmax,ss) and minimum (Cmin,ss)
concentrations can be used to compute clearance, volume of
Parameters in Patients distribution, and half-life. At steady state, consecutive Cmin,ss values
Foundation Issues

are equal, so the predose value can be extrapolated to the time


15 If it is necessary to determine the kinetic constants for a patient
before the next dose and used to calculate the half-life (dashed line).
to individualize his or her dose, a small kinetic evaluation is con-
ducted in the individual. In these cases, the number of serum con-
centrations obtained from the patient is held to the minimum needed
to calculate accurate pharmacokinetic parameters and doses. The
reason for using fewer serum drug concentration determinations is could result in a concentration too low for the assay to detect. In this
to be as cost-effective as possible because these laboratory tests gen- situation, the predose minimum and postdose maximum concentra-
erally cost $20 to $50 each. tions are used to compute V, where V = Dose/(Cmax − Cmin), and both
Although many drugs follow two-compartment-model pharma- postdose concentrations are used to calculate t1/2 (eFig. 5-11).
cokinetics (especially after IV administration), a one-compartment After CL, V, and t1/2 have been computed for a patient, the dose
model is used to compute kinetic parameters in patients because too and dosage interval necessary to achieve desired steady-state serum
many serum concentration determinations would be needed to deter- concentrations can be calculated using one-compartment-model
mine accurately both the distribution and elimination phases found in equations. Specific examples of these methods to calculate initial
the two-compartment model. Because of this, serum concentrations doses and individualized doses using serum concentrations are dis-
usually are not measured in patients during the distribution phase. cussed later in this chapter for the aminoglycoside antibiotics, van-
Another important reason serum concentrations are not measured comycin, digoxin, theophylline, phenytoin, and cyclosporine.
during the distribution phase for therapeutic drug-monitoring pur-
poses in patients is that drug in the blood and drug in the tissues are Computer Programs
not in equilibrium during this time, so that serum concentrations do Computer programs that aid in the individualization of therapy are
not reflect tissue concentrations. When drug serum concentrations are available for many different drugs. The most sophisticated programs
obtained in patients for the purpose of assessing efficacy or toxicity, it use nonlinear regression to fit CL and VD to actual serum concen-
is important that they be measured in the postdistribution phase when trations obtained in a patient.31 After drug doses and serum con-
drug in the blood is in equilibrium with drug at the site of action. centrations are entered into the computer, nonlinear least-squares
In the case where the patient has received enough doses to be regression programs adjust CL and VD until the sum of the squared
at steady state, pharmacokinetic parameters can be computed using error between actual (Cact) and computer-estimated concentrations
a predose minimum concentration and a postdose maximum con-
centration. Under steady-state conditions, serum concentrations
after each dose are identical, so the predose minimum concentra-
tion is the same before each dose (eFig. 5-10). This situation allows
10
the predose concentration to be used to compute both the patient’s Cmax
t1/2 and V, where V = Dose/(Cmax,ss − Cmin,ss). If the drug was given t1/2
extravascularly or has a significant distribution phase, the postdose
Concentration (mcg/mL)

concentration should be determined after absorption or distribution is


finished. To ensure that steady-state conditions have been achieved, C3
the patient needs to receive the drug on schedule for at least three to 1
Cmin
five estimated half-lives. To make sure that this is the case, inpatients
should have their medication administration records checked, and the
patient’s nurse should be consulted regarding missed or late doses.
Outpatients should be interviewed about compliance with the pre-
scribed dosage regimen. When compliance with the dosage regimen 0.1
0 1 2 3 4 5 6 7 8 9 10
has been verified, steady-state conditions reasonably can be assumed. Time (h)
If the patient is not at steady state, an additional postdose serum
concentration determination should be done to compute the patient’s eFigure 5-11  If a patient has not received enough doses to
pharmacokinetic parameters. Ideally, the third concentration (C3) be at steady state, or doses have been given on an irregular
should be acquired approximately one estimated half-life after the schedule, the minimum concentration (Cmin), maximum
postdose maximum concentration. Determining serum concentra- concentration (Cmax), and an additional postdose concentration
tions too close together will hamper the drug assay’s ability to mea- (C3) can be used to compute clearance, volume of distribution,
sure differences between them, and getting the third sample too late and half-life.
Copyright © 2014 McGraw-Hill Education. All rights reserved.
63
(Cest) is at a minimum [Σ(Cest - Cact)2]. Once estimates of CL and VD intermittent IV infusion equation at steady state, and the dose is
are available, doses are calculated easily. rounded off to the nearest 5 to 10 mg:
Many programs also take into account what the CL and VD 1 – e–kτ
should be on the basis of disease states and conditions present in the D = TkVDCmax,ss
1 – e–kT
patient.32 Incorporation of expected population-based parameters
allows the computer to use a limited number of serum concentra- The Hull and Sarrubi aminoglycoside dosage nomogram
tions (one or two) to provide estimates of CL and VD. This type of (eTable 5-5) is based on this dosage-calculation method and includes
precalculated doses and dosage intervals for a variety of creatinine

e|CHAPTER  
computer program is called Bayesian because it incorporates por-
tions of Bayes’ theorem during the fitting routine.33 Bayesian phar- clearance values.25 The nomogram assumes that VD = 0.26 L/kg and
macokinetic dosing programs are used widely to adjust the dose should not be used to compute doses for disease states with altered VD.
of a variety of drugs. In the case of renally eliminated drugs (e.g., An example of this initial dosage scheme for a typical case is
aminoglycosides, vancomycin, and digoxin), population estimates provided to illustrate the use of the various equations. Mr. JJ is a
for kinetic parameters are generated by entering the patient’s age,
weight, height, gender, and serum creatinine concentration into the
computer program. For hepatically eliminated drugs (e.g., theophyl-
line and phenytoin), population estimates for kinetic parameters are
eTable 5-5 Aminoglycoside Dosage Chart
1. Compute the patient’s creatinine clearance (CLcr) using the Cockcroft-
5
computed using the patient’s age, weight, and gender, as well as other Gault method: CLcr = [(140 – age)BW]/(Scr × 72) where Scr is expressed

Clinical Pharmacokinetics and Pharmacodynamics


factors that might change hepatic clearance, such as the presence in units of mg/dL. Multiply by 0.85 for women. (Scr expressed in μmol/L
must be divided by 88.4 to obtain conventional units of mg/dL.)
or absence of disease states (e.g., cirrhosis or CHF) or other drug
2. Use the patient’s weight if within 30% of IBW; otherwise use adjusted
therapy that might cause a drug interaction. The p­ opulation-based body weight ABW = IBW + [0.40(TBW – IBW)] where IBW and TBW are
estimates of the pharmacokinetic parameters are then modified expressed in kg.
using nonlinear least-squares regression fits of serum concentrations 3. Select the loading dose in mg/kg to provide peak serum concentrations
to result in individualized parameters for the patient. The individual- in the range listed below for the desired aminoglycoside antibiotic:
ized parameters are used to compute doses for the patient that will Expected Peak Serum
result in desired steady-state concentrations of the drug. Aminoglycoside Usual Loading Doses Concentrations
Tobramycin 1.5–2 mg/kg 4–10 mcg/mL or mg/L
9 to 21 μmol/L
Aminoglycosides Gentamicin 1.5–2 mg/kg 4 to 10 mcg/mL or mg/L
Although aminoglycoside pharmacokinetics follow multicom- 8 to 21 μmol/L
partment models,34 a one-compartment model appears sufficient Netilmicin 1.5–2 mg/kg 4 to 10 mcg/mL or mg/L
to individualize doses in patients.35 Aminoglycosides usually are 8 to 21 μmol/L
given as short-term intermittent IV infusions and administered as Amikacin 5–7.5 mg/kg 15–30 mcg/mL or mg/L
a single daily dose or multiple doses per day. Initial doses for ami- 26 to 51 μmol/L
noglycosides can be computed using estimated kinetic parameters Kanamycin 5–7.5 mg/kg 15 to 30 mcg/mL or mg/L
31 to 62 μmol/L
derived from population pharmacokinetic data. The elimination
rate constant is estimated using the patient’s creatinine clearance 4. Select the maintenance dose (as a percentage of the loading dose)
to continue peak serum concentrations indicated above according to
in the following formula: k (in h−1) = 0.00293(CLcr) + 0.014, where the desired dosage interval and the patient’s creatinine clearance. To
CLcr is the measured or estimated creatinine clearance in millili- maintain the usual peak/trough ratio, use dosage intervals in clear areas.
ters per minute. The volume of distribution is estimated using the
Percentage of Loading Dose Required for Dosage Interval Selected
average population value for normal-weight (within 30% of ideal
Estimated
weight) individuals equal to 0.26 L/kg [V = 0.26(Wt), where Wt CLcr (mL/min)b Half-Life (h) 8 h (%) 12 h (%) 24 h (%)
is the patient’s weight] or for obese individuals (>30% of ideal
>90 2–3 90 — —
weight)36 by taking into account the patient’s excess adipose tis-
90 3.1 84 — —
sue: V = 0.26[IBW + 0.4(TBW - IBW)], where TBW is total body
80 3.4 80 91 —
weight, IBW is ideal body weight [IBWmales (in kilograms) = 50 +
70 3.9 76 88 —
2.3(Ht - 60) or IBWfemales (in kilograms) = 45 + 2.3(Ht - 60), and Ht
is the patient’s height in inches] (height in cm can be converted to 60 4.5 71 84 —
inches by multiplying by 0.394). Additional volume of distribution 50 5.3 65 79 —
population estimates are available for other disease states and condi- 40 6.5 57 72 92
tions, such as cystic fibrosis,37 ascites,38 and neonates.39 30 8.4 48 63 86
Appropriate Cmax,ss and Cmin,ss values are selected for the patient 25 9.9 43 57 81
based on the site and severity of the infection and the sensitivity of 20 11.9 37 50 75
the known or suspected pathogen, as well as avoidance of adverse 17 13.6 33 46 70
effects. Optimal outcomes are usually associated with Cmax,ss/MIC 15 15.1 31 42 67
ratios equal to 8 to 10, where MIC is the minimum inhibitory con- 12 17.9 27 37 61
centration for the bacteria causing the infection. For example, Cmax,ss 10a 20.4 24 34 56
values of 8 to 10 mg/L (8 to 10 mcg/mL) generally are selected for  7a 25.9 19 28 47
gram-negative pneumonia patients, whereas Cmin,ss values of less than  5a 31.5 16 23 41
2 mg/L (2 mcg/mL; 4 μmol/L) usually are chosen to avoid amino-  2a 46.8 11 16 30
glycoside-induced nephrotoxicity when tobramycin and gentamicin
 0a 69.3  8 11 21
are prescribed using conventional multiple-daily-dosing regimens.
Once appropriate steady-state serum concentrations are selected, the ABW, adjusted body weight; BW, body weight; IBW, ideal body weight; Scr, serum
creatinine; TBW, total body weight.
dosage interval required to achieve those concentrations is calcu- a
Dosing for patients with CLcr ≤10 mL/min should be assisted by measuring serum
lated, and τ is rounded to a clinically acceptable value (e.g., 8, 12, concentrations.
18, 24, 36, or 48 hours): τ = [(ln Cmax,ss - ln Cmin,ss)/k] + T. Finally, a b
CLcr expressed in mL/min can be converted to units of mL/s by dividing by 60.
dose is computed for the patient using the one-compartment-model Data from Reference 25.

Copyright © 2014 McGraw-Hill Education. All rights reserved.


64
65-year-old, 80 kg (176 lb), 6-ft-tall (72 in. or 183 cm) man with If appropriate aminoglycoside serum concentrations are avail-
the diagnosis of gram-negative pneumonia. His serum creatinine able, kinetic parameters can be calculated at any point in therapy.
concentration is 2.1 mg/dL (186 μmol/L) and is stable. Compute When the patient is not at steady state, serum aminoglycoside con-
a conventional gentamicin dosage regimen (infused over 1 hour) centrations are obtained before a dose (Cmin), after a dose admin-
that would provide approximate peak and trough concentrations istered as an IV infusion of ∼1 hour or as a 30-minute infusion
of Cmax,ss = 8 mg/L (8 mcg/mL; 17 μmol/L) and Cmin,ss = 1.5 mg/L followed by a 30-minute waiting period to allow for drug distribu-
(1.5 mcg/mL; 3.1 μmol/L), respectively. The patient is within 30% tion (Cmax), and at one additional postdose time (C3) approximately
of his ideal body weight [IBWmale = 50 + 2.3(72 in - 60) = 78 kg] one estimated half-life after Cmax. The t1/2 and k values are computed
and has stable renal function, so the Cockcroft-Gault CLcr estimation using Cmax and C3: k = (ln Cmax - ln C3)/Δt and t1/2 = 0.693/k, where Δt
SECTION

equation can be used: CLcr est = [(140 - 65 y)80 kg]/[72(2.1 mg/dL)] = is the time that expired between the times Cmax and C3 were obtained.
40 mL/min (0.67 mL/s). The patient’s weight and estimated CLcr are If the patient is at steady state, serum aminoglycoside concentrations
used to compute his V and k, respectively: V = 0.26 L/kg(80 kg) = are obtained before a dose (Cmin,ss) and after a dose administered as
20.8  L; k  = 0.00293(40 mL/min) + 0.014 = 0.131  h−1 or t1/2 = an IV infusion of ∼1 hour or as a 30-minute infusion followed by
  

(0.693/0.131 h−1) = 5.3 h. The dosage interval and dose for the desired a 30-minute waiting period to allow for drug distribution (Cmax,ss).
1 serum concentrations would then be calculated: τ = [(ln 8 mg/L -
ln 1.5 mg/L)/0.131 h−1] + 1 h = 13.7 h rounded to 12 h; D = (1 h)
Because the patient is at steady state, it can be assumed that Cmin,ss is
identical for each dosage interval. The t1/2 and k values are computed
−1 −1
(0.131  h−1)(20.8 L)(8  mg/L) [1  - e−(0.131h (12h))/1 - e−(0.131h (1h))] = using Cmax,ss and Cmin,ss: k = (ln Cmax,ss - ln Cmin,ss)/(τ - T) and t1/2 =
Foundation Issues

140 mg. Thus, the prescribed dose would be gentamicin 140 mg every 0.693/k, where τ is the dosage interval, and T is the dose infusion
12 hours administered as a 1-hour infusion. If a loading dose were time or dose infusion time plus waiting time.
deemed necessary, it would be given as the first dose [LD = (20.8 L) Assuming a one-compartment model, the following equation is
(8 mg/L) = 166 mg rounded to 170 mg infused over 1 hour], and the used to compute VD:35
first maintenance dose would be administered 12  hours (e.g., one
dosage interval) later. Using the Hull and Sarrubi nomogram for the (D/T)(1 – e–kT)
VD =
same patient, the loading dose is 160 mg (gentamicin loading dose k(Cmax – Cmine–kT)
for serious gram-negative infection is 2 mg/kg: 2 mg/kg × 80 kg =
160 mg), and the maintenance dose is 115 mg every 12 hours (for a where D is the dose, and T is the duration of infusion. Once these
12-hour dosage interval and CLcr est = 40 mL/min (0.65 mL/s), main- are known, the dose and dosage interval (τ) can be calculated for any
tenance dose is 72% of the loading dose: 0.72 × 160 mg = 115 mg). desired maximum Css (Cmax,ss) and minimum Css (Cmin,ss):
For extended-interval therapy, Cmax,ss values of 20 to 30 mg/L
(20 to 30 mcg/mL; 42 to 63 μmol/L) and Cmin,ss values less than ln Cmax,ss – ln Cmin,ss
1 mg/L (1 mcg/mL; 2 μmol/L) generally are accepted as appropriate τ = +T
k
for gram-negative pneumonia patients. A minimum 24-hour dosage
1 – e–kτ
interval is chosen for this dosing technique, and the dosing inter- D = TkVDCmax,ss
val is increased in 12- to 24-hour increments for patients with renal 1 – e–kT
dysfunction.
The dose and dosage interval should be rounded to provide
An example of this initial dosage scheme for the same case is
clinically accepted values (every 8, 12, 18, 24, 36, and 48 hours for
provided to illustrate the use of extended-interval dosing. Mr. JJ is
dosage interval, nearest 5 to 10 mg for conventional dosing; every
65 years old, weighs 80 kg (176 lb). His height is 6 ft (72 in. [183 cm])
24, 36, and 48 hours for dosage interval, nearest 10 to 25 mg for
and his diagnosis is gram-negative pneumonia. His serum creatinine
extended interval dosing). This method also has been used to indi-
concentration is 2.1 mg/dL (186 μmol/L) and is stable. Compute an
vidualize IV theophylline dosage regimens.40
extended-interval gentamicin dosage regimen (infused over 1 hour)
To provide an example of this technique, the problem given
that would provide approximate peak and trough concentrations of
previously for conventional dosing will be extended to include
Cmax,ss = 25 mg/L (25 mcg/mL; 52 μmol/L) and Cmin,ss = 0.5 mg/L
steady-state concentrations. Please note that this method of dos-
(0.5 mcg/mL; 1 μmol/L), respectively. The patient is within 30%
age adjustment using serum concentrations can also be used for
of his ideal body weight [IBWmale = 50 + 2.3(72 in - 60) = 78 kg]
extended-interval dosing. Mr. JJ was prescribed gentamicin 140 mg
and has stable renal function, so the Cockcroft-Gault CLcr estima-
every 12 hours (infused over 1 hour) for the treatment of gram-neg-
tion equation can be used: CLcr est = [(140 - 65 y)80 kg]/[72(2.1 mg/
ative pneumonia. Steady-state trough (Cmin,ss) and peak (Cmax,ss) val-
dL)] = 40 mL/min (0.67 mL/s). The patient’s weight and estimated
ues were obtained before and after the fourth dose was given (more
CLcr are used to compute his V and k, respectively: V = 0.26 L/
than three to five estimated half-lives), respectively, and equaled
kg(80 kg) = 20.8 L; k = 0.00293(40 mL/min) + 0.014 = 0.131 h−1
Cmin,ss = 2.8 mg/L (2.8 mcg/mL; 5.9 μmol/L) and Cmax,ss = 8.5 mg/L
or t1/2 = (0.693/0.131 h−1) = 5.3 h. The dosage interval and dose for
(8.5  mcg/mL; 18 μmol/L). Clinically, the patient was improving
the desired serum concentrations would then be calculated: τ = [(ln
with decreased white blood cell counts and body temperatures and
25 mg/L - ln 0.5 mg/L)/0.131 h−1] + 1 h = 31 h rounded to 36 h; D =
−1 −1 a resolving chest radiograph. However, the serum creatinine value
(1 h)(0.131 h−1)(20.8 L)(25 mg/L) [1 - e−(0.131h (36h))/1 - e−(0.131h (1h))] =
had increased to 2.5 mg/dL (221 μmol/L). Because of this, a new
550 mg. Thus, the prescribed dose would be gentamicin 550 mg
dosage regimen with a similar peak (to maintain high intrapulmo-
every 36 hours administered as a 1-hour infusion.
nary levels) but lower trough (to decrease the risk of drug-induced
nephrotoxicity) concentrations was suggested. The patient’s elimi-
Clinical Controversy. . . nation rate constant and half-life can be computed using the fol-
lowing formulas: k = (ln 8.5 mg/L - ln 2.8 mg/L)/(12 h - 1 h) =
Some clinicians use conventional dosing or extended-interval
0.101 h−1 and t1/2 = 0.693/0.101 h−1 = 6.9 h. The patient’s volume of
dosing exclusively for patients requiring aminoglycosides,
distribution can be calculated using the following equation:
whereas others use a mix of both approaches according to
the perceived benefit to the patient. Definitive, authoritative
recommendations to guide the choice of one method of (140 mg/l h) 1 − e–(0.101 h–1)(1 h)
aminoglycoside dosing over the other are not available. V= = 22.3 L
(0.101 h–1) 8.5 mg/L − (2.8 mg/L)e–(0.101 h–1)(1 h)

Copyright © 2014 McGraw-Hill Education. All rights reserved.


65
Thus, the patient’s volume of distribution was larger and half-
life was longer than originally estimated; this led to higher serum 14
concentrations than anticipated. To achieve the desired serum con- 13
centrations (Cmin,ss = 1.5 mg/L [1.5 mcg/mL; 3.1 μmol/L] and Cmax,ss = 12
8 mg/L [8.0 mcg/mL; 17 μmol/L]), the patient’s actual kinetic param- 11

Concentration (mcg/mL)
eters are used to compute a new dose and dosage interval: τ = [(ln 10
8 mg/L - ln 1.5 mg/L)/0.101 h−1] + 1 h = 17.6 h, rounded to 18 h and 9
q 48 h

e|CHAPTER  
8

1 – e–(0.101 h–1)(18 h)
 7

D = (1 h)(0.101 h–1)(8 mg/L) 6 q 36 h

1 – e–(0.101 h–1)(1 h)

5

4 q 24 h
= 157 mg, round to 160 mg
3
Thus, the new dose would be gentamicin 160 mg every 18 2
hours and infused over 1 hour; the first dose of the new dosage regi-
men would be given 18 hours (e.g., the new dosage interval) after
6 7 8 9 10 11 12 13
Time between start of infusion and sample draw (h)
14
5
the last dose of the old dosage regimen. 1. Administer 7 mg/kg gentamicin or tobramycin with initial dosage

Clinical Pharmacokinetics and Pharmacodynamics


Because aminoglycoside antibiotics exhibit concentration- interval:
dependent bacterial killing, and the postantibiotic effect is longer
with higher concentrations, investigators studied the possibility of Estimated CLcr (mL/min) Initial dosage interval
giving a higher dose of aminoglycoside using an extended-dosage ≥60 mL/min q 24 h
interval (24 hours or longer, depending on renal function). Generally,
these studies have shown comparable microbiologic and clinical cure 40–59 mL/min q 36 h
rates for many infections and about the same rate of nephrotoxicity
20–39 mL/min q 48 h
(∼5-10%) as with conventional dosing. Ototoxicity has not been
monitored using audiometry in most of these investigations, but loss <20 mL/min Monitor serial concentrations
of hearing in the conversational range, as well as signs and symp- and administer next dose when
toms of vestibular toxicity, usually has been assessed and found to <1 mcg/mL
be similar to that with aminoglycoside therapy dosed conventionally.
Based on these data, clinicians are using extended-interval dosing in 2. Obtain timed serum concentration 6 to 14 hours after dose
selected patients. For Pseudomonas aeruginosa infections where the (ideally first dose)
organism has an expected minimum inhibitory concentration (MIC)
3. Alter dosage interval to that indicated by the nomogram
≈ 2 mg/L, peak concentrations between 20 and 30 mg/L (20 and zone (above q 48 h zone, monitor serial concentrations
30 mcg/mL) and trough concentrations less than 1 mg/L (1 mcg/mL; and administer next dose when <1 mcg/mL)
2 μmol/L) for gentamicin or tobramycin have been suggested.41
At the present time, there is no consensus on how to approach
concentration monitoring using this mode of administration. Some eFigure 5-12  Hartford nomogram for extended-interval
clinicians obtain steady-state peak and trough concentrations and aminoglycosides. (From Reference 41. Reproduced with permission
use the kinetic equations given earlier to adjust the dose and dosage from American Society for Microbiology.) To determine the
interval in order to attain appropriate target levels. Other clinicians corresponding creatinine clearance in units of mL/s divide the
measure only trough concentrations, trusting that the large doses value in mL/min by 60. Gentamicin levels in mcg/mL can be
administered to patients achieve adequate peak concentrations. converted to units of μmol/L by multiplying by 2.09.
Also, a nomogram that adjusts extended-interval doses based
on a single postdose concentration to achieve these Css goals has
been proposed (eFig. 5-12). The dose is 7 mg/kg of gentamicin
serum gentamicin concentration is 8.2 mg/L (17 μmol/L). According
or tobramycin. The initial dosage interval is set according to the
to the graph contained in the nomogram, the dosage interval should
patient’s CLcr. The Hartford nomogram includes a method to adjust
be changed to 48 hours. The new dose is 560 mg every 48 hours.
doses based on serum concentrations. This portion of the nomogram
contains average serum concentration time lines for gentamicin or
tobramycin in patients with creatinine clearances of 60, 40, and
20 mL/min (1, 0.67, 0.33 mL/s, respectively). A serum concentra-
Clinical Controversy. . .
tion is measured 6 to 14 hours after the first dose is given, and this “Trough only” measurement of steady-state vancomycin
concentration/time point is plotted on the graph (see eFig. 5-12). concentrations is a mainstream method to monitor
The modified dosage interval is indicated by which zone the serum therapy. The exact range for this value is uncertain. Many
concentration/time point falls. Because cystic fibrosis patients have clinicians recommend 5 to 15 mcg/mL (5 to 15 mg/L;
a different volume of distribution (0.35 L/kg) than assumed by this 3.45 to 10 μmol/L). For some sites of infection with specific
dosing technique, and extended-interval dosing has not been tested organisms, such as hospital-acquired pneumonia caused
adequately in patients with endocarditis, the Hartford nomogram by multidrug-resistant organisms, guidelines suggest
should not be used in these situations. vancomycin trough concentrations as high as 15 to 20 mcg/
To illustrate how the Hartford nomogram is used, the same mL (15 to 20 mg/L; 10 to 14 μmol/L) may be necessary. Some
patient example used previously will be repeated for this dos- clinicians continue to measure both steady-state peak and
age approach. Mr. JJ weighs 80 kg (176 lb) and has a CLcr est of trough vancomycin concentrations. Optimal outcomes are
40 mL/min (0.67 mL/s). Using the Hartford nomogram, the patient usually associated with AUC24/MIC ratios greater than 400,
would receive gentamicin 560 mg every 36 hours (7 mg/kg × where MIC is the minimum inhibitory concentration for the
80 kg = 560 mg; the initial dosage interval for CLcr est = 40 mL/min causative organism.
[0.67 mL/s] is 36 hours). Ten hours after the first dose was given, the
Copyright © 2014 McGraw-Hill Education. All rights reserved.
66
Vancomycin effects. Optimal outcomes are usually associated with AUC24/MIC
ratios greater than 400, where AUC24 is the area under the vancomy-
Vancomycin requires multicompartment models to completely cin concentration/time curve for 24 hours and MIC is the minimum
describe its serum-concentration-versus-time curves. However, if inhibitory concentration for the bacteria causing the infection. Cmax,ss
peak serum concentrations are obtained after the distribution phase values of between 20 and 40 mg/L (20 and 40  mcg/mL; 14 and
is completed (usually 30 minutes to 1 hour after a 1-hour IV infu- 28 μmol/L) and Cmin,ss values of between 5 and 15  mg/L (5 and
sion), a one-compartment model can be used for patient dosage 15 mcg/mL; 3 and 10 μmol/L) typically are used for patients with
calculations. Also, because vancomycin has a relatively long half- mild to moderate infections or sensitive bacteria with lower MIC
life compared with the infusion time, only a small amount of drug values (<1 mcg/mL). For patients with pneumonia or other life-­
SECTION

is eliminated during infusion, and it is usually unnecessary to use threatening infections due to multidrug-resistant organisms, Cmin,ss
more complex IV infusion equations. Thus, simple IV bolus equa- as high as 15–20 mg/L (15–20 mcg/mL; 10–14 μmol/L) have been
tions can be used to calculate vancomycin doses for most patients. suggested.48 After appropriate steady-state concentrations are cho-
Although a recent review article42 questioned the clinical usefulness sen, the dosage interval required to attain those concentrations is
  

of measuring vancomycin concentrations on a routine basis, other computed, and τ is rounded to a clinically acceptable value (8, 12,
1 research articles44,45 have shown potential benefits in obtaining van-
comycin concentrations in select patient populations. Some clini-
18, 24, 36, 48, or 72 hours): τ = (ln Cmax,ss - ln Cmin,ss)/k. Finally,
the maintenance dose is computed for the patient using a one-­
cians advocate monitoring only steady-state trough concentrations compartment-model IV bolus equation at steady state, and the dose
of vancomycin.46 The decision to conduct vancomycin concentra-
Foundation Issues

is rounded off to the nearest 100 to 250 mg:


tion monitoring should be made on a patient-by-patient basis.
Initial doses of vancomycin can be computed for adult patients D = Cmax,ss VD (1 - e-kt)
using estimated kinetic parameters derived from population phar- If desired, a loading dose can be computed using the following
macokinetic data. Clearance is estimated using the patient’s cre- equation:
atinine clearance in the following equation:43 CL (in mL/min/kg)
= 0.695(CLcr in mL/min/kg) + 0.05. The volume of distribution is LD = VD Cmax,ss
computed assuming the standard value of 0.7 L/kg: VD = 0.7(Wt), The following case will illustrate the use of this dosage meth-
where Wt is the patient’s weight. In the case of obese patients, actual odology. Ms. HJ is 65 years old, weighs 68 kg (150 lb), and is 5 ft
or total body weight is used in the calculation of clearance, but ideal 4  in. (64 in. [163 cm]) tall. She has developed a surgical wound
body weight is used to compute volume of distribution.47 The elim- infection; S. epidermidis is the suspected pathogen. Her serum cre-
ination rate constant is calculated using clearance and volume of atinine concentration is 1.8 mg/dL (159 μmol/L) and stable. Com-
distribution estimates, correcting for possible differences in units pute a vancomycin dosage regimen that would provide approximate
for these parameters: k = CL/VD. A nomogram that uses this type of peak (obtained 1 hour after a 1-hour infusion) and trough concen-
approach for vancomycin therapy is available to determine initial trations of 30 and 7 mg/L (30 and 7 mcg/mL; 21 and 5 μmol/L),
doses rapidly for patients (eTable 5-6).24 respectively. The patient is within 30% of her ideal body weight
Steady-state peak and trough concentrations are chosen for the [IBWfemale = 45 + 2.3(64 in. - 60) = 54 kg] and has stable renal
patient based on the site and severity of the infection, as well as function, so the Cockcroft-Gault creatinine clearance estimation
the known or suspected pathogen and avoidance of potential side formula can be used: CLcr est = 0.85[(140 - 65 y)68 kg]/[72(1.8 mg/
dL)] = 33 mL/min (0.55 mL/s). The patient’s weight and CLcr est are
used to calculate her estimated CL, VD, and k, respectively: CL =
eTable 5-6 Vancomycin Dosage Chart 0.695 (33 mL/min/68 kg) + 0.05 = 0.387 mL/min/kg; VD = 0.7 L/kg
1. Compute patient’s creatinine clearance (CLcr) using the Cockcroft-Gault (68 kg) = 48 L; and k = [(0.387 mL/min/kg)(68 kg)(60 min/h)]/
method: CLcr = [(140 – age)BW]/(SCr × 72), where Scr is expressed in units [(48 L)(1,000 mL/L)] = 0.033 h−1 or t1/2 = 0.693/0.033 h−1 = 21 h.
of mg/dL. Multiply by 0.85 for women. (Scr expressed in μmol/L must be The dosage interval, maintenance dose, and loading dose for the
divided by 88.4 to obtain conventional units of mg/dL.) desired serum concentrations can be computed: τ = (ln 30 mg/L -
2. Use the patient’s TBW to compute doses. ln 7 mg/L)/0.033 h−1 = 44 h, rounded to 48 h; D = (30 mg/L) (48 L)
3. Dosage chart designed to achieve peak serum concentrations of (1 - e−(0.033h−1)(48h)) = 1,145 mg, rounded to 1,250 mg; LD = (48 L)
30 mcg/mL (30 mg/L; 21 μmol/L) and trough concentrations of (30  mg/L) = 1,440 mg, rounded to 1,500 mg. Therefore, the pre-
7.5 mcg/mL (7.5 mg/L; 5 μmol/L).
scribed dose would be vancomycin 1,250 mg every 48 hours admin-
4. Compute loading dose of 25 mg/kg. istered as a 1-hour infusion. If a loading dose was used, it would
5. Compute maintenance dose of 19 mg/kg given at the dosage interval be given as the first dose, and the first maintenance dose would
listed in the following chart for the patient’s CLcr: be administered 48 hours (one dosage interval) later. Using the
CLcr (mL/min)a Dosage Interval (Days) Matzke nomogram for the same patient, the loading dose would
≥120 0.5 be 1,750  mg (vancomycin loading dose is 25 mg/kg: 25 mg/kg ×
 100 0.6
68  kg = 1,700  mg, rounded to 1,750 mg), followed by a mainte-
nance dose of 1,250 mg every 48 hours (for CLcr  est = 30 mL/min
  80 0.75
(0.50  mL/s)), the maintenance dose is 19 mg/kg every 2 days:
  60 1.0 19 mg/kg × 68 kg = 1,292 mg, rounded to 1,250 mg).
  40 1.5 If appropriate vancomycin serum concentrations are available,
  30 2.0 kinetic parameters can be computed at any point in therapy. When
  20 2.5 the patient is not at steady state, serum vancomycin concentrations
  10 4.0 are obtained before a dose (Cmin), after a dose administered as an IV
infusion of 1 hour followed by a 30-minute to 1-hour waiting period
   5 6.0
to allow for drug distribution (Cmax), and at one additional postdose
   0 12.0
time (C3) approximately one estimated half-life after Cmax. The t1/2
BW, body weight; Scr, serum creatinine; TBW = total body weight. and k values are computed using Cmax and C3: k = (ln Cmax - ln C3)/Δt
Clcr expressed in mL/min can be converted to units of mL/s by dividing by 60.
a
and t1/2 = 0.693/k, where Δt is the time that expired between the times
Data from Reference 24. Cmax and C3 were obtained. If the patient is at steady state, serum
Copyright © 2014 McGraw-Hill Education. All rights reserved.
67
vancomycin concentrations are obtained before a dose (Cmin,ss) and bacterial killing, the minimum concentration is the most important
after a dose administered as an IV infusion of approximately 1 hour with regard to therapeutic outcome. Vancomycin pharmacokinet-
followed by a 30-minute to 1-hour waiting period to allow for drug ics also support this approach because the volume of distribution is
distribution (Cmax,ss). The t1/2 and k values are computed using Cmax,ss relatively stable and is not changed by many disease states or condi-
and Cmin,ss: k = (ln Cmax,ss - ln Cmin,ss)/(τ - Tmax) and t1/2 = 0.693/k, tions. Because of this important point, it is difficult to attain peak
where τ is the dosage interval, and Tmax is the dose infusion time steady-state concentrations in the toxic range when the steady-state
plus waiting time. vancomycin trough is in the therapeutic range if typical doses are

e|CHAPTER  
Assuming a one-compartment model, the following equation is used (15 mg/kg or ≈ 1,000 mg for average-weight individuals). Also,
used to compute VD: toxic peak concentrations (generally greater than 80–100 mg/L [80 to
D 100 mcg/mL; 55 to 69 μmol/L]) are quite a bit higher than therapeu-
VD = tic peak concentrations, which adds a safety margin between effective
Cmax – Cmin
concentrations and those yielding adverse drug effects.
where D is dose. Once these are known, the dose and dosage inter- Coupled with trough-only vancomycin concentration monitor-
val (τ) can be calculated for any desired maximum Css(Cmax,ss) and ing is a widening of the therapeutic steady-state trough concentra-
minimum Css(Cmin,ss): tion range from 5 to 15 mg/L (5 to 15 mcg/mL; 3 to 10 μmol/L). The
justification for increasing the top of the range from 10 to 15 mg/L
5
ln Cmax,ss – ln Cmin,ss
τ= (10 to 15 mcg/mL; 7 to 10 μmol/L) comes from limited retrospec-

Clinical Pharmacokinetics and Pharmacodynamics


k tive45 and prospective45 studies. Trough concentrations in the range
D = Cmax,ssVD (1 – e–kτ ) of 15 to 20 mg/L (15 to 20 mcg/mL; 10 to 14 μmol/L) should be
reserved for specific clinical situations, such as hospital-acquired
The dose and dosage interval should be rounded to provide pneumonia or other severe infections caused by multidrug-resistant
clinically accepted values (every 8, 12, 18, 24, 36, 48, or 72 hours organisms.48 As previously mentioned, optimal outcomes are usu-
for dosage interval, nearest 100 to 250 mg for dose). ally associated with AUC24/MIC ratios greater than 400, where MIC
To provide an example for this dosage-calculation method, the is the minimum inhibitory concentration for the causative organism.
preceding problem will be extended to include steady-state con- When trough-only monitoring of vancomycin concentrations
centrations. Ms. HJ was prescribed vancomycin 1,200 mg every is chosen by a clinician, a simple variant of linear pharmacokinetics
48 hours (infused over 1 hour) for the treatment of a surgical wound can be used to adjust the dose (D) and dosage interval (τ): (Dnew/
infection. Steady-state trough (Cmin,ss) and peak (Cmax,ss) values τnew) = (Dold/τold)(Css,new/Css,old), where new and old indicate the new
(Cmax,ss obtained 1 hour after the end of the infusion) were obtained target trough concentration and the old measured trough concentra-
before and after the third dose was given (more than three to five tion, respectively. In practice, the dose (typically 1,000–1,500 mg)
estimated half-lives), respectively, and equaled Cmin,ss = 2.5 mg/L is often held constant, and only the dosage interval is changed. This
(2.5 mcg/mL; 1.7 μmol/L) and Cmax,ss = 22.4 mg/L (22.4 mcg/mL; equation is an approximation of the actual new steady-state trough
15.5 μmol/L). Clinically, the patient had improved somewhat, but concentration that will be attained in the patient because, mathemat-
her white blood cell count was still elevated, and the patient was still ically, Css,new is an exponential function of τ.
febrile. Because of this, a modified dosage regimen with a Cmax,ss = An example of this approach is given in the following case.
30 mg/L (30 mcg/mL; 21 μmol/L) and Cmin,ss = 7 mg/L (7 mcg/mL; Mr. MK is 72 years old, weighs 72 kg (158 lb), and measures 5 ft
5 μmol/L) was suggested to maintain an AUC24/MIC ratio greater 9 in. (69 in. [175 cm]). He was prescribed vancomycin 1,000 mg
than 400. The patient’s actual elimination rate constant and half-life every 12 hours (infused over 1 hour) for the treatment of an S. epi-
can be calculated using the following formulas: k = (ln 22.4 mg/L dermidis central venous catheter infection. A steady-state trough
- ln 2.5 mg/L)/(48 h - 2 h) = 0.048 h−1 and t1/2 = 0.693/0.048 h−1 = (Cmin,ss) value was obtained before the fifth dose was given (more than
14.4 h. The patient’s volume of distribution can be calculated using three to five estimated half-lives), and Cmin,ss = 19 mg/L (19 mcg/mL;
the following equation: 13  μmol/L). Clinically, the patient was improving, but the trough
concentration was judged to be too high. Because of this, a modified
1,200 mg
VD = = 60 L dosage regimen with a Cmin,ss = 10 mg/L (10 mcg/mL; 7 μmol/L) was
22.4 mg/L – 2.5 mg/L suggested to maintain an AUC24/MIC ratio greater than 400. (Dnew/
Thus, the patient’s volume of distribution was larger and τnew) = (1,000 mg/12 h)(10 mg/L/19 mg/L) = 44 mg/h. Because the
half-life shorter than originally estimated; this led to lower serum patient is near his ideal weight, the same dose of 1,000 mg can be
concentrations than anticipated. To achieve the desired serum con- used (Dnew), and the new dosage interval (τnew) can be computed:
centrations (Cmax,ss = 30 mg/L [30 mcg/mL; 21 μmol/L] and Cmin,ss = τ = 1,000 mg/44 mg/h = 23 h, rounded to 24 h. The new prescribed
7 mg/L [7 mcg/mL; 5 μmol/L]), the patient’s actual kinetic param- dose for the patient would be 1,000 mg every 24 hours.
eters are used to calculate a new dose and dosage interval:
ln 30 mg/L – ln 7 mg/L Digoxin
τ=
0.048 h –1 Digoxin pharmacokinetics are best described by a two-compartment
= 30 h, rounded to 36 h model. However, because digoxin has a long half-life compared
 
with its dosage interval and a very long distribution phase, simple
D = (30 mg/L)(60 L) 1 – e–(0.048 h–1)(36 h) pharmacokinetic equations can be used to individualize dosing
= 1,480 mg, rounded to 1,500 mg when postdistribution serum concentrations are used. Digoxin can
be given as an IV injection and orally as elixir (F = 0.8) or tablets
The new dose would be vancomycin 1,500 mg every 36 hours (F = 0.7). When given orally, the appropriate bioavailability fraction
(infused over 1 hour); the first dose of the new dosage regimen must be used to compute the correct dose. Initial doses of digoxin
would be given 36 hours (the new dosage interval) after the last dose can be computed using population pharmacokinetic data obtained
of the old dosage regimen. from published studies. Digoxin clearance is estimated using the
For routine monitoring, many clinicians measure only steady- patient’s CLcr in the following formula:23 CL (in milliliters per min-
state vancomycin trough concentrations in patients. The justification ute) = 1.303(CLcr in milliliters per minute)  + CLm, where CLm is
for this approach is that because vancomycin exhibits time-dependent metabolic clearance and equals 40 mL/min for patients with no or
Copyright © 2014 McGraw-Hill Education. All rights reserved.
68
mild heart failure or 20 mL/min for patients with moderate to severe computed by taking a detailed medical history of the patient and
heart failure. The volume of distribution decreases with declining noting disease states and conditions that are known to change the-
renal function and is estimated using the following equation:23 VD ophylline disposition. Age, smoking of tobacco-containing prod-
(in liters) = 226 + [298(CLcr in milliliters per minute)]/(29.1 + CLcr ucts, heart failure, and liver disease are among the important factors
in milliliters per minute). The elimination rate constant can be com- that alter theophylline kinetic parameters and dosage requirements.
puted by taking the product of CL and VD: k = CL/VD. For obese Once the patient has been assessed, average theophylline kinetic
individuals, digoxin dosing should be based on ideal body weight.49 parameters obtained from the literature for patients similar to the
Appropriate Css values are chosen for the patient based on the one being currently treated are used to compute either oral or IV
disease state being treated, the goal of therapy, and avoidance of doses. Dosage guidelines that take into account most common
SECTION

adverse effects. The inotropic effects of digoxin occur at lower con- disease states and conditions that change theophylline kinetic
centrations than do the chronotropic effects. Therefore, initial serum parameters are available (see eTable 5-4).52 Once theophylline is
concentrations of digoxin for the treatment of heart failure generally administered, the patient is monitored for the therapeutic effect and
are 1 ng/mL (1 mcg/L; 1.3 nmol/L) or less and for the treatment of potential adverse effects. Theophylline concentrations then are used
  

atrial fibrillation 1 to 1.5 ng/mL (1 to 1.5 mcg/L; 1.3 to 1.9 nmol/L). to individualize the theophylline dose that the patient receives. An
1 Once the appropriate Css is selected, a dose is computed for the
patient: D/τ = (CssCL)/F.
example of this approach was given previously for a patient in the
section on drug dosing in patients with liver disease (see Selection
An example of this initial dosage scheme is provided in the fol- of Initial Drug Doses above).
Foundation Issues

lowing case. Mr. PO is 72 years old, weighs 83 kg (183 lb), and mea- Continuous IV infusions of theophylline (or its salt, amino-
sures 5 ft 11 in. (71 in. [180 cm]). He was admitted to the hospital phylline) can be individualized rapidly by determining the patient’s
for the treatment of community-acquired pneumonia. His past medi- CL before steady state occurs.53 Assuming that the patient receives
cal history is positive for moderate heart failure. While in the hos- theophylline only by continuous IV infusion (previous doses of
pital, Mr. PO develops atrial fibrillation, and the decision is made to sustained-release oral theophylline are completely absorbed), two
treat him with digoxin to provide ventricular rate control. His serum serum theophylline concentration determinations are done 4 hours
creatinine concentration is 2.5 mg/dL (221 μmol/L) and stable. or more apart. The infusion rate (k0) cannot be changed between the
Calculate an IV loading dose and oral maintenance dose that will times the samples are drawn. With one-compartment model equa-
achieve a Css of 1.5 ng/mL (1.5 mcg/L; 1.9 nmol/L). The Cockcroft- tions, the first (C1) and second (C2) theophylline concentrations are
Gault equation can be used to estimate the patient’s CLcr because used to calculate theophylline CL:
his serum creatinine concentration is stable, and he is within 30% of
2k0 2VD(C1 – C2)
his ideal body weight [IBWmale = 50 + 2.3(71 in - 60) = 75 kg]: CLcr CL = +
= [(140 - 72 y)83 kg]/[72(2.5 mg/dL)] = 31 mL/min (0.52 mL/s). C1 + C2 (C1 + C2)(t2 – t1)
Using the estimated CLcr, both CL and VD can be computed:
VD is assumed to be 0.5 L/kg, and t1 and t2 are the times at
CL = 1.303(31 mL/min) + 20 = 60 mL/min which C1 and C2, respectively, are obtained. Once CL is known,
298(31 mL/min) k0 can be computed easily for any desired Css (Css = k0/CL). This
VD = 226 + = 380 L method probably can be applied to other drugs that are administered
29.1 + 31 mL/min
as continuous IV infusions, such as IV antiarrhythmics, when rapid
The maintenance dose will be given as digoxin tablets, so individualization of drug dosage is desirable.
F  = 0.7 in the dosing equation: D/τ = [(1.5 mcg/L)(60 mL/min) An example of this approach can be obtained by continuing the
(60 min/h)(24 h/day)]/[0.7(1,000 mL/L)] = 185 mcg/day, rounded to theophylline patient case from the section on drug dosing in liver
187.5 mcg/day (given as 1½ of 125 mcg tablets). The loading dose disease (see Selection of Initial Drug Doses above). In this example,
will be given IV as a digoxin injection: LD = (1.5 mcg/L)(380 L) a 55-year-old, 70 kg (154 lb) man with liver cirrhosis was prescribed
= 570 mcg, rounded to 500 mcg. The loading dose would be given a loading dose of theophylline 350 mg IV over 20 to 30 minutes, fol-
50% now (250 mcg), 25% (125 mcg) in 4 to 6 hours after moni- lowed by a maintenance dose of 15 mg/h of theophylline as a con-
toring the patient’s heart rate and blood pressure and assessing the tinuous infusion. The infusion began at 9 am, blood samples were
patient for digoxin adverse effects, and the final 25% (125 mcg) 4 to obtained at 10 am and 4 pm, and the clinical laboratory reported the
6 hours later after monitoring the same clinical parameters. The first theophylline serum concentrations as 10.9 and 12.3 mg/L (10.9 and
maintenance dose would be given one dosage interval (in this case, 12.3 mcg/mL; 60.5 and 68.3 μmol/L), respectively. The patient’s
24 hours) after the first part of the loading dose was given. theophylline clearance and revised continuous infusion to maintain
Adjustment of digoxin doses using steady-state concentrations a Css of 15 mg/L (15 mcg/mL; 83.3 μmol/L) can be computed as
is accomplished using linear pharmacokinetics and dosage ratios: follows (patient’s VD estimated at 0.5 L/kg):
Dnew = Dold(Css,new/Css,old). For example, Mr. PO’s atrial fibrillation
responded to digoxin therapy, and he was discharged after resolution 2(15 mg/h)
CL =
of his pneumonia. A month later, he was followed up in the clinic with 10.9 mg/L + 12.3 mg/L
moderate nausea, possibly a result of digoxin toxicity. His heart rate 2(0.5 L/kg × 70 kg)(10.9 mg/L – 12.3 mg/L)
was 51 beats per minute, and his serum creatinine was unchanged. A + = 0.59 L/h
(10.9 mg/L + 12.3 mg/L)(16 – 10 h)
steady-state digoxin concentration was determined and reported by
the clinical laboratory as 2.2 mcg/L (2.8 nmol/L). Compute a new dose k0 = CssCL = (15 mg/L)(0.59 L/h) = 9 mg/h theophylline
for the patient to achieve a Css of 1.5 mcg/L (1.9 nmol/L). The digoxin
Css and old dose would be used to calculate a new dose using the If theophylline is to be given as the aminophylline salt form,
linear pharmacokinetic equation: Dnew = 187.5 mcg/day[(1.5 mcg/L)/ the doses would need to be changed to reflect the fact that amino-
(2.2 mcg/L)] = 128 mcg/day, rounded to 125 mcg/day. phylline contains only 85% theophylline (k0 = 9 mg/h theophylline/
0.85 = 11 mg/h aminophylline).
If continuous IV infusions or oral dosage regimens are given
Theophylline long enough for steady state to occur (three to five estimated half-
Theophylline disposition is described most accurately by nonlinear lives based on previous studies conducted in similar patients), lin-
kinetics.50,51 However, at the usual doses, theophylline acts as if it ear pharmacokinetics can be used to adjust doses for either route
obeys linear kinetics in most patients. Initial theophylline doses are of administration: Dnew = Dold(Css,new/Css,old). For example, a patient
Copyright © 2014 McGraw-Hill Education. All rights reserved.
69
receiving 200 mg of sustained-release oral theophylline every
Vmax
12  hours with a theophylline steady-state serum concentration of
9.5 mcg/mL (9.5 mg/L; 52.7 μmol/L) can have the dose required to
achieve a new Css equal to 15 mcg/mL (15 mg/L; 83.3 μmol/L) com-
puted by applying linear pharmacokinetics: Dnew = 200 mg[(15 mcg/
mL)/(9.5 mcg/mL)] = 316 mg, rounded to 300 mg. Thus the new
Slope = −Km
theophylline dose would be 300 mg every 12 hours.

e|CHAPTER  
DR
Phenytoin
Phenytoin doses are very difficult to individualize because the drug
follows Michaelis-Menten kinetics, and there is a large amount of
interpatient variability in Vmax and Km. Initial maintenance doses
of phenytoin in adults usually range between 4 and 7 mg/kg daily,
yielding starting doses of 300 to 400 mg daily in most individuals.
If needed, loading doses of phenytoin or fosphenytoin (a prodrug DR/Css
5
of phenytoin used IV) can be administered in adults at a dose of

Clinical Pharmacokinetics and Pharmacodynamics


15 mg/kg, which is approximately 1,000 mg in many individu- eFigure 5-13  Relationship between dosage rate (DR) and
als. Loading doses of phenytoin can be given orally but need to be steady-state serum concentrations (Css).
administered in divided doses separated by several hours in order to
avoid decreased bioavailability and gastrointestinal intolerance (for
total loading dose of 1,000 mg: 400 mg, 300 mg, then 300 mg with 1.15(12.3 mcg/mL) = 14.1 mcg/mL or Cnew = 1.33 (12.3 mcg/mL) =
each dose separated by 4 to 6 h). Because phenytoin is metabolized 16.4 mcg/mL]. Thus, the patient would be expected to have a steady-
hepatically, decreased doses may be needed in patients with liver state phenytoin concentration of approximately 14 to 16  mcg/mL
disease. Because phenytoin follows dose-dependent pharmacokinet- (14 to 16 mg/L; 56 to 63 μmol/L) as a consequence of the dosage
ics, the half-life of phenytoin increases for a patient as the mainte- increase. An alternative approach would be to use a graphic Bayes-
nance dose increases. Therefore, the time to steady-state phenytoin ian method that allows an estimate of Vmax and Km from one steady-
concentrations increases with dose. On average, at a phenytoin dose state phenytoin concentration and the prediction of new steady-state
of 300 mg daily, it takes approximately 5 to 7 days to achieve steady concentrations when doses are changed.55
state; at a dose of 400 mg daily, it takes approximately 10 to 14 days Other methods used to individualize phenytoin doses involve
to achieve steady state; and at a dose of 500 mg daily, it takes approx- rearrangements of the Michaelis-Menten equation [DR = VmaxCss/
imately 21 to 28 days to achieve steady state. It should be noted that (Km + Css), in which DR is the dosage rate at steady state] so that two
the injectable and capsule dosage forms of phenytoin are phenytoin or more doses and Css values can be used to obtain graphic solutions
sodium, and the labeled dosage amounts contain 92% of active for Vmax and Km. One rearrangement56 is DR = -Km(DR/Css) + Vmax.
phenytoin (300 mg phenytoin sodium capsules contain 276  mg When DR is plotted on the y axis, and DR/Css is plotted on the x axis
[300 mg × 0.92 = 276 mg] of active phenytoin). Unbound phenytoin of Cartesian graph paper, a straight line with a y intercept of Vmax
concentrations are useful in patients with hypoalbuminemia (e.g., and slope equal to -Km is found (eFig. 5-13). To use this method,
liver disease, nephrotic syndrome, pregnancy, cystic fibrosis, burns, patients are prescribed an initial phenytoin dose, and Css is obtained.
trauma, and malnourishment, as well as in the elderly), in patients in The phenytoin dose is then changed, and a second Css from the new
whom displacement with endogenous compounds is possible (e.g., dose is obtained. Each dose is divided by its respective Css to derive
hyperbilirubinemia, liver disease, or end-stage renal disease), and DR/Css values. The DR/Css and Css values are plotted on the graph to
in patients receiving other drugs that may displace phenytoin from calculate Vmax (y intercept) and Km (minus slope). The steady-state
plasma protein-binding sights (e.g., valproic acid, aspirin therapy Michaelis-Menten equation can be used to compute Css.
>2 g daily, warfarin, and nonsteroidal antiinflammatory drugs with
high albumin binding).54
After steady state has occurred, phenytoin serum concentra- Cyclosporine
tions can be obtained as an aid to dosage adjustment. A simple, easy Because of the large amount of variability in cyclosporine pharma-
way to approximate new serum concentrations after a dosage adjust- cokinetics, even when concurrent disease states and conditions are
ment with phenytoin is to temporarily assume linear pharmacoki- identified, many clinicians believe that the use of standardized initial
netics and then add 15% to 33% for a dosage increase or subtract cyclosporine doses for various situations is warranted. Indeed, most
15% to 33% for a dosage decrease to account for Michaelis-Menten transplant centers use doses that are determined employing a locally
kinetics. To avoid large disproportionate changes in phenytoin con- derived cyclosporine dosage protocol. The original computations
centrations when using this empirical method, dosage adjustments of these doses were based on the pharmacokinetic dosing methods
should be limited to 50 to 100 mg daily. This technique is intended described in preceding sections and subsequently modified based on
only to provide a rough approximation of the resulting phenytoin Css clinical experience. In general, the expected cyclosporine Css used to
after an appropriate dosage adjustment has been made. compute these doses depends on the type of transplanted tissue and
For example, Ms. PP is a 35-year-old, 65 kg (143 lb) patient the posttransplantation time line. Generally speaking, initial oral
with grand mal seizures who is receiving phenytoin capsules 300 mg doses of 8 to 18 mg/kg daily or IV doses of 3 to 6 mg/kg daily (one-
orally at bedtime. A Css of 9.2 mcg/mL (9.2 mg/L; 37 μmol/L) is third the oral dose to account for ∼30% oral bioavailability) are used
measured. It is observed that her seizure frequency decreased by and vary greatly from institution to institution. For obese individuals
only ∼15%, and that she has had no adverse effects as a conse- (>30% over ideal body weight), ideal body weight should be used to
quence of phenytoin treatment. Because of this, her phenytoin dose compute initial doses.
is increased to 400 mg orally at bedtime. The expected phenytoin Css It is likely that doses computed using patient population
would be estimated using linear pharmacokinetics [Cnew = (Dnew/Dold) characteristics will not always produce cyclosporine concentra-
Cold = (400 mg/300 mg)/(9.2 mcg/mL) = 12.3 mcg/mL] and then tions that are expected or desirable. Additionally, there is a very
increased by 15% to 33% to account for nonlinear kinetics [Cnew = high amount of interday variation in cyclosporine concentrations.
Copyright © 2014 McGraw-Hill Education. All rights reserved.
70
eTable 5-7 Recommended 2-hour (±15 min) Postdose CLINICAL PHARMACODYNAMICS
Steady-state Cyclosporine Concentrations
(C2) for Various Solid Organ Transplant 16 Pharmacodynamics is the study of the relationship between
Types and Posttransplant Times the concentration of a drug and the response obtained in a patient.
Renal Transplant
Originally, investigators examined the dose–response relationship
posttransplant Time (Months) C2 Level
of drugs in humans but found that the same dose of a drug usually
resulted in different concentrations in individuals because of phar-
1 1,500–2,000 ng/mL or mcg/L
1,248-1,664 nmol/L macokinetic differences in clearance and volume of distribution.
Examples of quantifiable pharmacodynamic measurements include
SECTION

2 1,500 ng/mL or mcg/L


1,248 nmol/L changes in blood pressure during antihypertensive drug therapy,
decreases in heart rate during β-blocker treatment, and alterations in
3 1,300 ng/mL or mcg/L
1,082 nmol/L prothrombin time or international normalized ratio during warfarin
therapy.
  

4–6 1,100 ng/mL or mcg/L


915 nmol/L For drugs that exhibit a direct and reversible effect, the follow-
1 7–12 900 ng/mL or mcg/L
ing diagram describes what occurs at the level of the drug receptor:
749 nmol/L
Drug + receptor ↔ drug - receptor complex ↔ response
>12 800 ng/mL or mcg/L
Foundation Issues

666 nmol/L According to this scheme, there is a drug receptor located


Liver Transplant within the target organ or tissue. When a drug molecule “finds” the
Posttransplant Time (Months) C2 Level receptor, it forms a complex that causes the pharmacologic response
0–3 1,000 ng/mL or mcg/L to occur. The drug and receptor are in dynamic equilibrium with the
832 nmol/L drug–receptor complex.
4–6 800 ng/mL or mcg/L
666 nmol/L
The Emax and Sigmoid Emax Models
>6 600 ng/mL or mcg/L
499 nmol/L The mathematical model that comes from the classic drug receptor
theory shown previously is known as the Emax model:
Data from References 57, 58, and 59.
Emax × C
E=
EC 50 + C
Because of pharmacokinetic variability, the narrow therapeutic
index of cyclosporine, and the severity of cyclosporine adverse side where E is the pharmacologic effect elicited by the drug, Emax is
effects, measurement of cyclosporine concentrations is mandatory the maximum effect the drug can cause, EC50 is the concentration
for patients to ensure that therapeutic, nontoxic levels are present. causing one-half the maximum drug effect (Emax/2), and C is the
When cyclosporine concentrations are measured in patients, and a concentration of drug at the receptor site. EC50 can be used as a mea-
dosage change is necessary, clinicians should seek to use the sim- sure of drug potency (a lower EC50, indicating a more potent drug),
plest, most straightforward method available to determine a dose whereas Emax reflects the intrinsic efficacy of the drug (a higher
that will provide safe and effective treatment. In most cases, a sim- Emax, indicating greater efficacy). If pharmacologic effect is plotted
ple dosage ratio can be used to change cyclosporine doses using against concentration in the Emax equation, a hyperbola results with
steady-state concentrations and assuming that the drug follows lin- an asymptote equal to Emax (eFig. 5-14). At a concentration of zero,
ear pharmacokinetics: no measurable effect is present.
When dealing with human studies in which a drug is adminis-
Css,new tered to a patient, and pharmacologic effect is measured, it is very
Dnew = Dold
Css,old difficult to determine the concentration of the drug at the recep-
tor site. Because of this, serum concentrations (total or unbound)
The Css can be either a steady-state trough concentration or a
Css measured 2 hours (±15 min) after a dose (C2). When C2 levels
are used, recommended concentrations vary according to transplant
type and posttransplant time (see eTable 5-7).57–59 100
For example, LK is a 50-year-old, 75 kg (165 lb), 5 ft 11 in.
(71  in. [180 cm]) male renal transplant recipient who is receiv-
80
ing oral cyclosporine 400 mg every 12 hours. The current steady-
state blood cyclosporine concentration is 375 ng/mL (375 mcg/L;
312  nmol/L). To compute a cyclosporine dose that will provide a 60
Css of 200 ng/mL (200 mcg/L; 166 nmol/L), linear pharmacokinetic
Effect

equations can be used. The new dose to attain the desired concentra-
40
tion should be proportional to the old dose that produced the mea-
sured concentration (total daily dose = 400 mg/dose × 2 doses/d =
800 mg/d): 20

Css,new 200 ng/mL


Dnew = Dold = 800 mg/day
Css,old 375 ng/mL
0 50 100 150 200 250 300 350
= 427 mg/day, round to 400 mg/day Concentration

The new suggested dose would be 400 mg/day or 200 mg every eFigure 5-14  The Emax model [E = (Emax × C)/(EC50 + C)] has the
12 hours of cyclosporine capsules to be started at the next scheduled shape of a hyperbola with an asymptote equal to Emax. EC50 is the
dosing time. concentration where effect = Emax/2.
Copyright © 2014 McGraw-Hill Education. All rights reserved.
71
usually are used as the concentration parameter in the Emax equation. 80
Therefore, the values of Emax and EC50 are much different than if
the drug were added to an isolated tissue contained in a laboratory
beaker. 60
The result is that a much more empirical approach is used to
describe the relationship between concentration and effect in clini-
cal pharmacology studies. After a pharmacodynamic experiment

Effect
40

e|CHAPTER  
has been conducted, concentration–effect plots are generated. The
shape of the concentration–effect curve is used to determine which
pharmacodynamic model will be used to describe the data. Because
20
of this, the pharmacodynamic models used in a clinical pharmacol-
ogy study are deterministic in the same way that the shape of the
serum-concentration-versus-time curve determines which pharma-
cokinetic model is used in clinical pharmacokinetic studies. 0 10 20 30 40 50 60 70 80
Sometimes a hyperbolic function does not describe the
­concentration–effect relationship at lower concentrations adequately.
Concentration 5
When this is the case, the sigmoid Emax equation may be supe- eFigure 5-16  The linear model (E = S × C + I) is often used as a

Clinical Pharmacokinetics and Pharmacodynamics


rior to the Emax model: pharmacodynamic model when the measured pharmacologic
effect is 20% to 80% of Emax. In this situation, the determination
Emax × C n
E= of Emax and EC50 is not possible. To illustrate this, effect
EC n50 + C n measurements from eFigure 5-14 between 20% and 80% of Emax
are graphed using the linear pharmacodynamic model.
where n is an exponent that changes the shape of the ­concentration–
effect curve. When n > 1, the concentration–effect curve is S- or
sigmoid-shaped at lower serum concentrations. When n < 1, the
­concentration–effect curve has a steeper slope at lower concentra-
because lower drug concentrations may not be detectable with the
tions (eFig. 5-15).
analytic technique used to assay serum samples, and higher drug
With both the Emax and sigmoid Emax models, the largest
concentrations may be avoided to prevent toxic side effects. The
changes in drug effect occur at the lower end of the concentra-
equation used is that of a simple line: E = S × C + I, where E is the
tion scale. Small changes in low serum concentrations cause large
drug effect, C is the drug concentration, S is the slope of the line, and
changes in effect. As serum concentrations become larger, further
I is the y intercept. In this situation, the value of S can be used as a
increases in serum concentration result in smaller changes in effect.
measure of drug potency (the larger the value of S, the more potent
Using the Emax model as an example and setting Emax = 100 units
the drug). The linear model can be derived from the Emax model.
and EC50 = 20 mg/L, doubling the serum concentration from 5 to
When EC50 is much greater than C, E = (Emax/EC50)C = S × C, where
10 mg/L increases the effect from 20 to 33 units (a 67% increase),
S = Emax/EC50.
whereas doubling the serum concentration from 40 to 80 mg/L only
The linear model allows a nonzero value for effect when the
increases the effect from 67 to 80 units (a 19% increase). This is an
concentration equals zero. This may be a baseline value for the
important concept for clinicians to remember when doses are being
effect that is present without the drug, the result of measurement
titrated in patients.
error when determining effect, or model misspecification. Also, this
model does not allow the prediction of a maximum response.
Linear Models Some investigators have used a log-linear model in pharma-
When serum concentrations obtained during a pharmacodynamic codynamic experiments: E = S × (log C) + I, where the symbols
experiment are between 20% and 80% of Emax, the concentration– have the same meaning as in the linear model. The advantages
effect curve may appear to be linear (eFig. 5-16). This occurs often of this model are that the concentration scale is compressed on
­concentration–effect plots for experiments where wide concentra-
tion ranges were used, and the concentration values are transformed
so that linear regression can be used to compute model parameters.
100
The disadvantages are that the model cannot predict a maximum
effect or an effect when the concentration equals zero. With the
increased availability of nonlinear regression programs that can
80 compute the parameters of nonlinear functions such as the Emax
model easily, use of the log-linear model has been discouraged.60
60
Baseline Effects
Effect

40 At times, the effect measured during a pharmacodynamic study


has a value before the drug is administered to the patient. In these
cases, the drug changes the patient’s baseline value. Examples of
20
these types of measurements are heart rate and blood pressure. In
addition, a given drug may increase or decrease the baseline value.
Two basic techniques are used to incorporate baseline values into
0 50 100 150 200 250 300 350
Concentration
pharmacodynamic data. One way incorporates the baseline value
into the pharmacodynamic model; the other transforms the effect
eFigure 5-15  The sigmoid Emax model [E = (Emax × Cn)/(ECn50 + Cn)] data to take baseline values into account.
has an S-shaped curve at lower concentrations. In this example, Incorporation of the baseline value into the pharmacodynamic
Emax and EC50 have the same values as in eFigure 5-14. model involves the addition of a new term to the previous equations.
Copyright © 2014 McGraw-Hill Education. All rights reserved.
72
E0 is the symbol used to denote the baseline value of the effect that 80
will be measured. The form that these equations takes depends
70
on whether the drug increases or decreases the pharmacodynamic
effect. When the drug increases the baseline value, E0 is added to 60
the equations:
50
Emax × C
E = E0 +

Effect
EC50 + C 40

Emax × C n
SECTION

30
E = E0 +
EC n50 + C n
20
E = S × C + E0
10
When E0 is not known with any better certainty than any other
  

effect measurement, it should be estimated as a model parameter


1 similar to the way that one would estimate the values of Emax, EC50,
S, or n.61,62 If the baseline effect is well known and has only a small
0 20 40 60 80
Concentration
100 120 140

amount of measurement error, it can be subtracted from the effect eFigure 5-17  Hysteresis occurs when effect measurements
Foundation Issues

determined in the patient during the experiment and not estimated are different at the same concentration. This is commonly
as a model parameter. This approach can lead to better estimates seen after short-term IV infusions or extravascular doses
of the remaining model parameters.62 Using the linear model as an where concentrations increase and subsequently decrease.
example, the equation used would be E - E0 = S × C. Counterclockwise hysteresis loops are found when
If the drug decreases the baseline value, the drug effect is sub- concentration–effect points are joined as time increases (shown
tracted from E0 in the pharmacodynamic models: by arrows) and effect is larger at the same concentration but
Emax × C at a later time. Clockwise hysteresis loops are similar, but the
E = E0 – concentration–effect points are joined in clockwise order, and
IC50 + C
the effect is smaller at a later time.
Emax × C n
E = E0 –
IC n50 + C n
E = E0 – S × C Hysteresis
where Emax represents the maximum reduction in effect caused by Concentration–effect curves do not always follow the same pat-
the drug, and IC50 is the concentration that produces a 50% inhibi- tern when serum concentrations increase as they do when serum
tion of Emax. These forms of the equations have been called the inhib- concentrations decrease. In this situation, the concentration–effect
itory Emax and inhibitory sigmoidal Emax equations, respectively. In curves form a loop that is known as hysteresis. With some drugs, the
this arrangement of the pharmacodynamic model, E0 is a model effect is greater when serum concentrations are increasing, whereas
parameter and can be estimated. If the baseline effect is well known with other drugs, the effect is greater while serum concentrations are
and has little measurement error, the effect in the presence of the decreasing (eFig. 5-17). When individual concentration–effect pairs
drug can be subtracted from the baseline effect and not estimated as are joined in time sequence, this results in clockwise and counter-
a model parameter. Using the inhibitory Emax model as an example, clockwise hysteresis loops.
the formula would be E0 - E = (Emax × C)/(IC50 + C). Clockwise hysteresis loops usually are caused by the devel-
When using the inhibitory Emax model, a special situation occurs opment of tolerance to the drug. In this situation, the longer the
if the baseline effect can be obliterated completely by the drug (e.g., patient is exposed to the drug, the smaller is the pharmacologic
decreased premature ventricular contractions during antiarrhythmic effect for a given concentration. Therefore, after an extravascular
therapy). In this situation, Emax = E0, and the equation simplifies to a or short-term infusion dose of the drug, the effect is smaller when
rearrangement known as the fractional Emax equation: serum concentrations are decreasing compared with the time
when serum concentrations are increasing during the infusion or
E = E0 1 – C  absorption phase.
 IC50 + C  Accumulation of a drug metabolite that acts as an antagonist
also can cause clockwise hysteresis.
This form of the model relates drug concentration to the frac- Counterclockwise hysteresis loops can be caused by the accu-
tion of the maximum effect. mulation of an active metabolite, sensitization to the drug, or delay
An alternative approach to the pharmacodynamic modeling in time in equilibration between serum concentration and concentra-
of drugs that alter baseline effects is to transform the effect data tion of drug at the site of action. Combined pharmacokinetic/phar-
so that they represent a percentage increase or decrease from the macodynamic models have been devised that allow equilibration lag
baseline value.62 For drugs that increase the effect, the following times to be taken into account.
transformation equation would be used: percent effectt = [(treat-
mentt - baseline)/baseline] × 100. For drugs that decrease the
effect, the following formula would be applied to the data: percent
inhibitiont = [(baseline - treatmentt)/baseline] × 100. The subscript
SUMMARY
indicates the treatment, effect, or inhibition that occurred at time The availability of inexpensive, rapidly achievable serum drug con-
t during the experiment. If the study included a placebo control centrations has changed the way clinicians monitor drug therapy in
phase, baseline measurements made at the same time as treatment patients. The therapeutic range for many drugs is known, and it is
measurements (i.e., heart rate determined 2 hours after placebo likely that more drugs will be monitored using serum concentra-
and 2 hours after drug treatment) could be used in the appropriate tions in the future. Clinicians need to remember that the therapeu-
transformation equation.62 The appropriate model (excluding E0) tic range is merely an average guideline and to take into account
then would be used. interindividual pharmacodynamic variability when treating patients.
Copyright © 2014 McGraw-Hill Education. All rights reserved.
73
Individual patients may respond to smaller concentrations or require 8. Gibaldi M, Koup JR. Pharmacokinetic concepts: Drug
concentrations that are much greater to obtain a therapeutic effect. binding, apparent volume of distribution and clearance.
Conversely, patients may show toxic effects at concentrations within Eur J Clin Pharmacol 1981;20:299–305, PMID:7308282.
or below the therapeutic range. Serum concentrations should never 9. Bowdle TA, Patel IH, Levy RH, et al. Valproic acid dosage
replace clinical judgment. and plasma protein binding and clearance. Clin Pharmacol
Three kinetic constants determine the dosage requirements of Ther 1980;28:486–492, PMID:6773716.
patients. Clearance determines the maintenance dose (MD = CLCss), 10. Lima JJ, Boudonlas H, Blanford M. Concentration-

e|CHAPTER  
volume of distribution determines the loading dose (LD = VDCss), dependence of disopyramide binding to plasma protein and
and half-life determines the time to steady state and the dosage its influence on kinetics and dynamics. J Pharmacol Exp
interval. Several methods are available to compute these parameters. Ther 1981;219:741–747, PMID:7299695.
Methods available to individualize drug therapy range from 11. Bertilsson L, Höjer B, Tybring G, et al. Autoinduction of
clinical pharmacokinetic techniques using simple mathematical carbamazepine metabolism in children examined by a stable
relationships that hold for all drugs that obey linear pharmaco- isotope technique. Clin Pharmacol Ther 1980;27(1):83–88,
kinetics to very complex computer programs that are specific to PMID:7351120.
one drug. 12. Gibaldi M, Perrier D. Pharmacokinetics. 2nd ed. New York:
Marcel Dekker, 1980.
5
13. Gibaldi M. Estimation of the pharmacokinetic parameters
ABBREVIATIONS

Clinical Pharmacokinetics and Pharmacodynamics


of the two-compartment open model from post-infusion
plasma concentration data. J Pharm Sci 1969;58:1133–1135,
ALT alanine aminotransferase
PMID:5346080.
AST aspartate aminotransferase
14. Loo JCK, Riegelman S. Assessment of pharmacokinetic
AUC area under the concentration-versus-time curve
constants from postinfusion blood curves obtained after
CHF chronic heart failure
IV infusion. J Pharm Sci 1970;59:53–55, PMID:5411325.
CL clearance
15. Wagner JG. Model-independent linear pharmacokinetics.
CLcr creatinine clearance
Drug Intell Clin Pharm 1976;10:179–180.
CLcr est estimated creatinine clearance
16. Hansten PD, Horn JR. The Top 100 Drug Interactions:
CLR renal clearance
A Guide to Patient Management. Freeland, WA: H&H
Cmax maximum serum or blood concentrations
Publications, 2013.
Css steady-state drug concentration
17. Giacomini KM, Huang SM, Tweedie DJ, et al. Membrane
CYP cytochrome P450
transporters in drug development. Nat Rev Drug Discov
D dose
2010;15(3):215–236.
DR dosage rate
18. Zhang L, Huang SM, Lesko LJ. Transporter-mediated
Emax the maximum pharmacologic effect elicited by a drug
drug-drug interactions. Clin Pharmacol Ther 2011;89(4):
GI gastrointestinal
481–484.
ka absorption rate constant
19. Cockcroft DW, Gault MH. Prediction of creatinine
LD loading dose
clearance from serum creatinine. Nephron 1976;16:31–41,
MIC minimum inhibitory concentration
PMID:1244564.
P-gp P-glycoprotein
20. Traub SL, Johnson CE. Comparison of methods of
t1/2 half-life
estimating creatinine clearance in children. Am J Hosp
VD volume of distribution
Pharm 1980;37:195–201, PMID:7361791.
Vmax maximum rate of metabolism
21. Salazar DE, Corcoran GB. Predicting creatinine clearance
and renal drug clearance in obese patients from estimated
REFERENCES fat-free body mass. Am J Med 1988;84:1053–1060,
PMID:3376975.
1. Koup JR, Gibaldi M. Some comments on the evaluation 22. Jelliffe RW, Jelliffe SM. A computer program for estimation
of bioavailability data. Drug Intell Clin Pharm 1980;14: of creatinine clearance from unstable serum creatinine levels,
327–330. age, sex, and weight. Math Biosci 1972;14:17–24.
2. Gibaldi M, Boyes RN, Feldman S. Influence of first pass 23. Koup JR, Jusko WJ, Elwood CM, Kohli RK. Digoxin
effect on availability of drugs on oral administration. pharmacokinetics: Role of renal failure in dosage
J Pharm Sci 1971;60:1338–1340, PMID:5567579. regimen design. Clin Pharmacol Ther 1975;18:9–21,
3. Wu C-Y, Benet LZ, Hebert MF, et al. Differentiation of PMID:1149366.
absorption and first-pass gut and hepatic metabolism in 24. Matzke GR, McGory RW, Halstenson CE, Keane WF.
humans: Studies with cyclosporine. Clin Pharmacol Ther Pharmacokinetics of vancomycin in patients with various
1995;58:492–497, PMID:7586942. degrees of renal function. Antimicrob Agents Chemother
4. Wagner JG, Northam JI, Alway CD, et al. Blood levels of 1984;25:433–437, PMID:6732213.
drug at the equilibrium state after multiple dosing. Nature 25. Sarubbi FA, Hull JH. Amikacin serum concentrations:
1965;207:1301–1302, PMID:5884654. Predictions of levels and dosage guidelines. Ann Intern Med
5. Rowland M, Benet LZ, Graham GG. Clearance concepts in 1978;89:612–618, PMID:717929.
pharmacokinetics. J Pharmacokinet Biopharm 1973;1: 26. Sivan SK, Bennett WM. Drug dosing guidelines in patients
123–136, PMID:4764426. with renal failure. West J Med 1992;156:633–638.
6. Wilkinson GR, Shand DG. A physiological approach to 27. Aronoff GR, Bennett WM, Berns JS, et al. Drug Prescribing
hepatic drug clearance. Clin Pharmacol Ther 1975;18: in Renal Failure. 5th ed. Philadelphia, PA: American College
377–390, PMID:1164821. of Physicians, 2007.
7. Nies AS, Shand DG, Wilkinson GR. Altered hepatic blood 28. Pugh RNH, Murray-Lyon IM, Dawson JL, et al. Transection
flow and drug disposition. Clin Pharmacokinet 1976;1: of the oesophagus for bleeding oesophageal varices. Br J
135–155, PMID:13954. Surg 1973;60:646–649, PMID:4541913.
Copyright © 2014 McGraw-Hill Education. All rights reserved.
74
29. Jusko WJ, Gardner MJ, Mangione A, et al. Factors affecting 46. Karam CM, McKinnon PS, Neuhauser MM, Rybak MJ.
theophylline clearances: Age, tobacco, marijuana, cirrhosis, Outcome assessment of minimizing vancomycin monitoring
congestive heart failure, obesity, oral contraceptives, and dosage adjustments. Pharmacotherapy 1999;19:257–266,
benzodiazepines, barbiturates, and ethanol. J Pharm Sci PMID:10221365.
1979;68:1358–1366, PMID:41932. 47. Blouin RA, Bauer LA, Miller DD, et al. Vancomycin
30. Thomson PD, Melmon KL, Richardson JA, et al. Lidocaine pharmacokinetics in normal and morbidly obese subjects.
pharmacokinetics in advanced heart failure, liver disease, and Antimicrob Agents Chemother 1982;21:575–580,
renal failure in humans. Ann Intern Med 1973;78:499–508, PMID:7081978.
PMID:4694036. 48. Niederman MS, Craven DE. Guidelines for the management
SECTION

31. Koup JR, Killen T, Bauer LA. Multiple-dose nonlinear of adults with hospital-acquired, ventilator-associated, and
regression analysis program: Aminoglycoside dose prediction. healthcare-associated pneumonia. Am J Respir Crit Care
Clin Pharmacokinet 1983;8:456–462, PMID:6627810. Med 2005;171(4):388–416.
32. Sheiner LB, Beal S, Rosenberg B, et al. Forecasting 49. Abernethy DR, Greenblatt DJ, Smith TW. Digoxin disposition
  

individual pharmacokinetics. Clin Pharmacol Ther in obesity: Clinical pharmacokinetic investigations. Am Heart
1 1979;26:294–305, PMID:466923.
33. Sheiner LB, Beal SL. Bayesian individualization of
J 1981;102:740–744, PMID:7282520.
50. Sarrazin E, Hendeles L, Weinberger M, et al. Dose-
pharmacokinetics: Simple implementation and comparison dependent kinetics for theophylline: Observations among
Foundation Issues

with non-Bayesian methods. J Pharm Sci 1982;71: ambulatory asthmatic children. J Pediatr 1980;97:825–828,
1344–1348, PMID:7153881. PMID:7431179.
34. Schentag JJ, Jusko WJ. Renal clearance and tissue 51. Tang-Liu DDS, Williams RL, Riegelman S. Nonlinear
accumulation of gentamicin. Clin Pharmacol Ther 1977; theophylline elimination. Clin Pharmacol Ther 1982;31:
22:364–370, PMID:330081. 358–369, PMID:7060318.
35. Sawchuk RJ, Zaske DE, Cipolle RJ, et al. Kinetic model 52. Edwards DJ, Zarowitz BJ, Slaughter RL. Theophylline.
for gentamicin dosing with the use of individual patient In: Evans E, Schentag JJ, Jusko WJ, eds. Applied
parameters. Clin Pharmacol Ther 1977;21:362–369, Pharmacokinetics: Principles of Therapeutic Drug Monitoring.
PMID:837654. Vancouver, WA: Applied Therapeutics, 1992:13–1-13–38.
36. Bauer LA, Edwards WAD, Dellinger EP, Simonowitz DA. 53. Vozeh S, Kewitz G, Wenk M, et al. Rapid prediction of
Influence of weight on aminoglycoside pharmacokinetics steady-state serum theophylline concentrations in patients
in normal weight and morbidly obese patients. Eur J Clin treated with intravenous aminophylline. Eur J Clin
Pharmacol 1983;24:643–647, PMID:6873144. Pharmacol 1980;18:473–477, PMID:7461015.
37. Bauer LA, Piecoro JJ, Wilson HD, Blouin RA. Gentamicin 54. Bauer LA. Use of mixed-effect modeling to determine the
and tobramycin pharmacokinetics in patients with cystic influence of albumin, bilirubin, valproic acid, warfarin, and
fibrosis. Clin Pharm 1983;2:262–264, PMID:6883955. aspirin on phenytoin unbound fraction and pharmacokinetics.
38. Sampliner R, Perrier D, Powell R, Finley P. Influence of J Am Pharm Assoc 2004;44:236–237.
ascites on tobramycin pharmacokinetics. J Clin Pharmacol 55. Vozeh S, Muir KT, Sheiner LB, Follath F. Predicting
1984;24:43–46, PMID:6707233. individual phenytoin dosage. J Pharmacokinet Biopharm
39. Zank KE, Miwa L, Cohen JL, et al. Effect of body weight 1991;9:131–146.
on gentamicin pharmacokinetics in neonates. Clin Pharm 56. Ludden TM, Allen JP, Valutsky WA, et al. Individualization
1984;3:170–173. of phenytoin dosage regimens. Clin Pharmacol Ther
40. Pancorbo S, Sawchuk RJ, Dashe C, et al. Use of a 1977;21:287–293, PMID:837647.
pharmacokinetic model for individual intravenous doses of 57. Cole E, Midtvedt K, Johnston A, et al. Recommendations
aminophylline. Eur J Clin Pharmacol 1979;16:251–254. for the implementation of Neoral C(2) monitoring in clinical
41. Nicolau DP, Freeman CD, Belliveau PP, et al. Experience practice. Transplantation 2002;73(9 suppl):S19–S22.
with a once-daily aminoglycoside program administered 58. Levy G, Thervet E, Lake J, et al. Patient management
to 2184 adult patients. Antimicrob Agents Chemother by Neoral C(2) monitoring: An international consensus
1995;39:650–655, PMID:7793867. statement. Transplantation 2002;73(9 suppl):S12–S18.
42. Cantu TG, Yamanaka-Yuen NA, Lietman PS. Serum 59. Levy GA. C2 monitoring strategy for optimising cyclosporine
vancomycin concentrations: Reappraisal of their clinical immunosuppression from the Neoral formulation. BioDrugs
value. Clin Infect Dis 1994;18:533–543, PMID:8038306. 2001;15(5): 279–290, PMID:11437692.
43. Moellering RC Jr, Krogstad DJ, Greenblatt DJ. Vancomycin 60. Holford NHG, Sheiner LB. Understanding the dose-effect
therapy in patients with impaired renal function: A relationship: Clinical application of pharmacokinetic-
nomogram for dosage. Ann Intern Med 1981;94:343–346, pharmacodynamic models. Clin Pharmacokinet 1981;6:
PMID:6101256. 429–453, PMID:7032803.
44. Welty TE, Copa AK. Impact of vancomycin therapeutic 61. Schwinghammer TL, Kroboth PD. Basic concepts
drug monitoring on patient care. Ann Pharmacother in pharmacodynamic modeling. J Clin Pharmacol
1994;28:1335–1339, PMID:7696720. 1988;28:388–394, PMID:3292599.
45. Zimmermann AE, Katona BG, Plaisance KI. Association 62. Sheiner LB, Stanski DR, Vozeh S, et al. Simultaneous
of vancomycin serum concentrations with outcomes in modeling of pharmacokinetics and pharmacodynamics:
patients with gram-positive bacteremia. Pharmacotherapy Application to d-tubocurarine. Clin Pharmacol Ther
1995;15:85–91, PMID:7739950. 1979;25:358, PMID:761446.

Copyright © 2014 McGraw-Hill Education. All rights reserved.

You might also like